MXPA06001231A - Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with an anticholinergic bronchodilator for treatment of asthma or chronic obstructive pulmonary disease. - Google Patents

Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with an anticholinergic bronchodilator for treatment of asthma or chronic obstructive pulmonary disease.

Info

Publication number
MXPA06001231A
MXPA06001231A MXPA06001231A MXPA06001231A MXPA06001231A MX PA06001231 A MXPA06001231 A MX PA06001231A MX PA06001231 A MXPA06001231 A MX PA06001231A MX PA06001231 A MXPA06001231 A MX PA06001231A MX PA06001231 A MXPA06001231 A MX PA06001231A
Authority
MX
Mexico
Prior art keywords
dhea
alkyl
adenosine
pharmaceutically acceptable
pharmaceutical composition
Prior art date
Application number
MXPA06001231A
Other languages
Spanish (es)
Inventor
Cynthia B Robinson
Original Assignee
Epigenesis Pharmaceuticals Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Epigenesis Pharmaceuticals Llc filed Critical Epigenesis Pharmaceuticals Llc
Publication of MXPA06001231A publication Critical patent/MXPA06001231A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Pulmonology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Rheumatology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Immunology (AREA)
  • Pain & Pain Management (AREA)
  • Neurosurgery (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Steroid Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

A pharmaceutical or veterinary composition, comprises a first active agent selected from a dehydroepiandrosterone and/or dehydroepiandrosterone-sulfate, or a salt thereof, and a second active agent comprising an anticholinergic bronchodilator for the treatment of asthma, chronic obstructive pulmonary disease, or other respiratory diseases. The composition is provided in various formulations and in the form of a kit. The products of this patent are applied to the prophylaxis and treatment of asthma, chronic obstructive pulmonary disease, or other respiratory diseases.

Description

COMBINATION OF DEHYDROEPIANDROSTERONE OR DEHYDROEPIANDROSTERONE SULFATE WITH A BRONCODILATOR ANTICOLINÉRGICO FOR THE TREATMENT OF ASTHMA OR CHRONIC OBSTRUCTIVE PULMONARY DISEASE FIELD OF THE INVENTION The present invention relates to a composition comprising a non-glucocorticoid steroid including dehydroepiandrosterone (DHEA), DHEA sulfate, or a salt thereof, and an anticholinergic bronchodilator. These compositions are useful in the treatment of asthma, chronic obstructive pulmonary disease (COPD), or other respiratory diseases. BACKGROUND OF THE INVENTION Respiratory ailments, associated with a variety of conditions, are extremely common in the general population. In some cases they are accompanied by inflammation, which aggravates the condition of the lungs. Respiratory conditions include asthma, chronic obstructive pulmonary disease (COPD), and other respiratory diseases of the upper and lower airways, such as allergic rhinitis, acute respiratory distress syndrome (ARDS), and pulmonary fibrosis. Asthma, for example, is one of the most common diseases in industrialized cities. In the United States, it represents approximately 1% of total health care costs. There has been an alarming increase in both the frequency and mortality of asthma in the past decade and it is predicted that asthma will be the pre-eminent occupational disease of the lung in the next decade. Asthma is a condition characterized by a variable, in many cases reversible, obstruction of the airways. This process is associated with pulmonary inflammation and in some cases pulmonary allergies. Many patients have acute episodes referred to as "asthma attacks" while others are affected with a chronic condition. It is believed that the asthmatic process is caused in some cases by the inhalation of antigens by hypersensitive subjects. This condition is generally referred to as "extrinsic bronchial asthma". Other asthmatics have an intrinsic predisposition to the condition, which is referred to as "intrinsic asthma" and may include conditions of different origin, including those mediated by the adenosine receptors, allergic conditions mediated by an immune response mediated by IgE , and others'. All asthmatics have a group of symptoms, which are characteristic of this condition: episodic bronchoconstriction, pulmonary inflammation and decreased pulmonary surfactant. Existing bronchodilators and anti-inflammatories are currently commercially available and are prescribed for the treatment of asthma. The most common anti-inflammatories, corticosteroids, have considerable side effects but are nevertheless commonly prescribed. Most of the drugs available for the treatment of asthma are, in an important way, scarcely effective in many patients. COPD is characterized by obstruction of airflow that is usually caused by chronic bronchitis, emphysema, or both. Commonly, airway obstruction is incompletely reversible but 10 to 20% of patients show some improvement in airway obstruction with treatment. In chronic bronchitis, obstruction of the airways results from a chronic and excessive secretion of abnormal mucus in the airways, inflammation, bronchospasm and infection. Chronic bronchitis is also characterized by chronic cough, mucus production, or both, for at least three months in at least two successive years where other causes of chronic cough have been excluded. In emphysema, a structural element (elastin) in the terminal bronchioles is destroyed, leading to the collapse of the airway walls and the inability to exhale "stale" air. In emphysema there is a permanent destruction of the alveoli. Emphysema is characterized by abnormal permanent elongation of distal air spaces to the terminal bronchioles, accompanied by the destruction of its walls and without obvious fibrosis. COPD can also lead to secondary pulmonary hypertension. Secondary pulmonary hypertension itself is a disorder in which the blood pressure in the pulmonary arteries is abnormally high. In severe cases, the right side of the heart must work harder than usual to pump blood against high blood pressure. If this continues for a long period, the right side of the heart enlarges and works poorly, and collects fluids in the ankles (edema) and belly. Eventually the left side of the heart begins to fail. Heart failure caused by lung disease is called cor pulmonale. COPD characteristically affects middle-aged and elderly people, and is one of the leading causes of morbidity and mortality worldwide. In the United States it affects around 14 million people and is the fourth cause of death, and the third cause of disability in the United States of America. Both, morbidity and mortality, however, are growing. The estimated incidence of this disease in the United States of America has increased to 41% since 1982, and the death rates with respect to age rose to 71% between 1966 and 1985. This contrasts with the decrease in the same period in mortality with respect to age for all causes of death (which fell to 22%), and cardiovascular diseases (which fell to 45%). In 1998, COPD caused 112,584 deaths in the United States. COPD, however, is preventable, since it is believed that its main cause is exposure to cigarette smoke. Long-term smoking is the most common cause of COPD. It represents 80 to 90% of all cases. A smoker is 10 times more likely than a non-smoker to die from COPD. The disease is rare in the lives of non-smokers, in whom exposure to environmental tobacco smoke will explain at least some of the obstructions of the airways. Other proposed etiological factors include hyper-responsiveness or hypersensitivity of the airways, environmental air pollution, and allergy. Obstruction of the airways in COPD is usually progressive in people who continue to smoke. This results in early disability and reduced survival time. Quitting smoking shows the rate of deterioration of a non-smoker but the damage caused by smoking is irreversible. Other risk factors include: inheritance, passive smoking, exposure to polluted air at work and in the environment, and a history of respiratory infections during childhood. Symptoms of COPD include: chronic cough, chest tightness, inter-cut breathing at rest and during exercise, increased effort to breathe, increased production of mucus, and frequent throat clearing. There is very little currently available to alleviate the symptoms of COPD, prevent exacerbations, maintain optimal lung function, and improve daily activities and quality of life. Many patients will use medications chronically for the rest of their lives, with the need for increased doses and additional drugs during exacerbations. Medications that are currently prescribed for patients with COPD include: rapid-acting β2-agonists, anticholinergic bronchodilators, long-acting bronchodilators, antibiotics, and expectorants. Among the currently available treatments for COPD, there are short-term benefits, but no long-term effects, in their progress, from the administration of anticholinergic drugs, β2-adrenergic agonists and oral steroids. Oral steroids are only recommended for acute exacerbations with long-term use that contribute to increased mortality and morbidity. Long-acting and short-acting inhaled β2-adrenergic agonists achieve short-term bronchodilation and provide some relief of symptomatology in patients with COPD, but do not show any significant supportive effect on the progress of the disease. Β2 adrenergic agonists improve symptoms in subjects with COPD, such as an increased ability to exercise and produce some degree of bronchodilation, and even an increase in lung function in some severe cases. The maximum effectiveness of the most recent long-acting inhaled adrenergic β2-adrenergic agonists was found to be comparable to that of the fast-acting β2 adrenergic agonsites. It was discovered that Salmeterol improves symptomatology and quality of life, although it only produces modest changes or no changes in lung function. The use of p2-agonists can produce cardiovascular effects, such as changes in pulse rate, blood pressure and electrocardiogram results. In rare cases, the use of P2-agonists can produce hypersensitivity reactions, such as urticaria, angioedema, rash and oropharyngeal edema. In these cases, the use of the p2-agonist must be interrupted. The continuous treatment of asthmatic and COPD patients with bronchodilators such as ipratropium bromide or fenoterol was not superior to treatment on the basis of as required, thus indicating that they are not appropriate for maintenance treatment. The most common immediate adverse effect of β2-adrenergic agonists, on the other hand, is tremors, which in high doses can cause a drop in plasma potassium, dysrhythmias, and reduced blood pressure of oxygen. The combination of a β2 adrenergic agonist with an anticholinergic drug provides additional small bronchodilation compared with either drug alone. The addition of ipratropium to a standard dose of inhaled P2-adrenergic agonists for about 90 days, however, produces some improvement in stable patients with COPD with respect to any of the drugs alone. In general, the presence of adverse effects with P2-adrenergic agonists, such as tremors and dysrhythmias, is more frequent than with anticholinergics. Thus, neither anticholinergic drugs nor β2-adrenergic agonists have an effect on all people with COPD; nor the two agents combined. Anticholinergic drugs achieve short-term bronchodilation-and produce some relief of symptomatology in people with COPD, but not a better long-term prognosis. Most patients with COPD have at least some degree of airway obstruction that is somehow relieved by ipratropium bromide. The Lung Health Study found spirometric signs of early COPD in male and female smokers and followed up for five years. Three treatments were compared over a period of five years and the results showed that ipratropium bromide had no significant effect on the decrease in the effective functional volume of the patients' lungs whereas quitting caused a delay in the decrease in volume functional cash of the lungs. Ipratropium bromide, however, produced adverse effects, such as cardiac symptoms, hypertension, skin rash, and urinary retention. Theophyllines produce modest bronchodilation in patients with COPD although they produce frequent adverse effects and a small therapeutic interval. Serum concentrations of 15-20 mg / 1 are required to obtain optimal effects and serum levels must be carefully verified. Adverse effects include nausea, diarrhea, headache, irritability, convulsions, and cardiac arrhythmias, occurring at very variable blood concentrations and, in many people, still within the therapeutic range. Dosages of theophyllines should be adjusted individually according to smoking habits, infection, and other treatments, which are uncomfortable. Although theophyllines have been claimed to have an anti-inflammatory effect in asthma, especially at low doses, none have been reported in COPD. The adverse effects of theophyllines and the need to verify them frequently restrict their usefulness. Oral corticosteroids have been shown to improve the short-term outcome in acute exacerbations of COPD but the long-term administration of oral steroids has been associated with serious side effects including osteoporosis and inducing overt diabetes. It has been found that inhaled corticosteroids do not have a real short-term effect on hypersensitivity of the airways to histamine. In two 3-year treatment studies with inhaled fluticasone, moderate and severe exacerbations were significantly reduced as well as a modest improvement in quality of life without affecting lung function. Patients who suffer from COPD with a more reversible disease seem to benefit more from treatment with inhaled fluticasone. Mucolytics have a modest beneficial effect on the frequency and duration of exacerbations but an adverse effect on lung function. Neither acetylcysteine nor other mucolytics, however, have a significant effect in people with severe COPD (effective functional volume <50%) despite evidencing greater reductions in the frequency of exacerbation. N-acetylcysteine produced gastrointestinal side effects.
Long-term oxygen therapy given to patients with hypoxemic COPD and patients with congestive heart failure had little effect on their mortality rates during the first 500 days or so, but survival rates in men subsequently increased and remained constant for the next five years. In women, however, oxygen decreased mortality rates during the study. Continuous treatment with oxygen for patients with hypoxemic COPD during 19.3 years decreased the total risk of death. To date, however, it has been found that only lifestyle changes, smoking cessation, and long-term oxygen therapy (in hypoxemics) alter the long-term course of COPD. Antibiotics are often also administered at the first signs of respiratory infection to prevent further damage and infection in diseased lungs. Expectorants help loosen and expel mucous secretions from the airways, and can help facilitate breathing. In addition, other medications may be prescribed to control the conditions associated with COPD. These could include: diuretics (which are given as therapy to prevent excessive water retention associated with right heart failure), digitalis (which invigorates the strength of the heartbeat), and cough suppressants. This last list of medications helps relieve symptoms associated with COPD but does not treat COPD. Thus, there is very little currently available to alleviate the symptoms of COPD, avoid exacerbations, preserve optimal lung function, and improve daily life activities and quality of life. Acute Respiratory Distress Syndrome (ARDS), or rigid lung, lung in shock, pump lung, and congestive atelectasis, are thought to be caused by the buildup of fluid within the lung, which in turn causes the lung to put on rigid. The condition is triggered within 48 hours by a variety of processes that injure the lungs such as trauma, head injury, shock, sepsis, multiple blood transfusions, medications, pulmonary embolism, severe pneumonia, smoke inhalation, radiation, high altitude , being close to drowning, and others. In general, ARDS occurs as a medical emergency and may be caused by other conditions that directly or indirectly cause the fluid in the blood vessels to "leak" into the lungs. In ARDS, the ability of the lungs to expand is severely diminished and they cause extensive damage to the air sacs and the lining or endothelium of the lungs. The most common symptoms of ARDS are rapid, forced breathing, nasal widening, skin, lips, and cyanotic blue nails caused by lack of oxygen to the tissues, anxiety, and temporary absence of breathing. A preliminary diagnosis of ARDS can be confirmed with X-rays of the chest and with measurement of arterial blood gas. In some cases, ARDS appears to be associated with other diseases, such as acute myelogenous leukemia, with acute tumor lysis syndrome (SLTA) developed after treatment with, e.g. cytosine arabinoside. In general, however, ARDS seems to be associated with traumatic injury, severe blood infections such as sepsis, or other systemic diseases, high-dose radiation therapy and chemotherapy, and inflammatory responses that lead to multiple organ failure, and in many cases death. In premature babies ("preemies"), neither the lung tissue nor the surfactant are fully developed. When respiratory distress syndrome (RDS) occurs in preemies, it is an extremely serious problem. Preterm infants who show SRD are currently treated by ventilation and administration of oxygen and surfactant preparations. When preemies survive RDS, they frequently develop bronchopulmonary dysplasia (BPD), also called chronic lung disease of early childhood, which is often fatal. Allergic rhinitis affects one in five Americans, corresponding to an estimated $ 4-10 trillion in health care costs per year, and occurs at any age. Because many people do not identify their symptoms such as persistent colds or sinus problems, allergic rhinitis is probably misdiagnosed. Typically, IgE combines with allergens in the nose to produce chemical mediators, induction of cellular processes and neurogenic stimulation, which causes underlying inflammation. Symptoms include nasal and ocular congestion, nasal discharge, sneezing, and itching. At the time, patients suffering from allergic rhinitis often develop sinusitis, otitis media with effusion, and nasal polyposis. Approximately 60% of patients with allergic rhinitis also have asthma and the flashes of allergic rhinitis aggravate asthma. The degranulation of mast cells results in the release of preformed mediators that interact with various cells, blood vessels, and mucous glands to produce typical rhinitis symptoms. Most reactions of early and late phases occur in the nose after exposure to the allergen. The late phase reaction is seen in chronic allergic rhinitis, with hypersecretion and congestion as the most prominent symptoms. Repeated exposure causes a hypersensitivity reaction to one or many allergens. Patients can also become hyperreactive to non-specific factors such as cold air or strong odors. Non-allergic rhinitis can be induced by infections, such as viral infections, or associated with nasal polyps, as occurs in patients with idiosyncrasy to aspirin. Medical conditions such as pregnancy or hypothyroidism and exposure to occupational factors or medications can cause rhinitis. The so-called NARES syndrome (Non-allergic rhinitis with Eosinophilia syndrome) is a type of non-allergic rhinitis associated with eosinophils in nasal secretions, which typically occurs in middle-aged patients and is accompanied by some loss of sense of smell. The treatment of allergic and nonallergic rhinitis is not satisfactory. Self-administration of saline improves nasal obstruction, sneezing, and congestion and generally does not cause side effects and is therefore the first treatment applied in pregnant patients. Saline aerosols are generally used to relieve mucosal irritation or dryness associated with various nasal disorders, minimize mucosal atrophy, and dislodge thick, crusted mucus. If used immediately before doses of intranasal corticosteroids, saline aerosols can help avoid local irritation induced by medications. Antihistamines such as terfenadine and astemizole are also used to treat allergic rhinitis; however, the use of antihistamines has been associated with a ventricular arrhythmia known as Torsades de Points, generally in interaction with other medications such as guetoconazole and erythromycin or secondary to an underlying cardiac problem. Loratadine, another non-sedating antihistamine and cetirizine have not been associated with an adverse impact in the QT interval, or with serious adverse cardiovascular events. Cetirizine, however, produces extreme drowsiness and has not been widely prescribed. Non-sedating antihistamines, e. g. claritin, may produce some relief from sneezing, runny, nasal, ocular and palatal itching, but it has not been tested for asthma or other more specific conditions. Terfenadine, loratadine and astemizole, on the other hand, show extremely modest bronchodilator effects, reduction of bronchial hyperresponsiveness to histamine, and protection against bronchospasm induced by antigens and exercise. Some of these benefits, however, require higher doses than currently recommended. Sedative antihistamines help induce sleep at night, but cause drowsiness and compromise performance if taken during the day. Antihistamines, when used, are typically combined with a decongestant to help improve nasal congestion. Sympathomimetic drugs are used as vasoconstrictors and decongestants. The three commonly prescribed systemic decongestants, pseudoephedrine, phenylpropanolamine and phenylephrine, cause hypertension, palpitations, tachycardia, restlessness, insomnia and headache. The interaction of phenylpropanolamine with caffeine in doses of two or three cups of coffee can significantly raise blood pressure. In addition, medications such as pseudoephedrine can cause hyperactivity in children. Topical decongestants, however, are indicated only for a limited period of time, as they are associated with a rebound nasal dilatation when used in excess. Anticholinergic agents are provided to patients with significant rhinorrhea or for specific conditions such as "gustatory rhinitis", usually caused by ingestion of spicy foods, and may have some beneficial effect on the common cold. Chromolin, for example, if used prophylactically as a nasal spray, reduces sneezing, runny nose, and nasal itching, and blocks sensitive responses in both early and late stages, but produces sneezing, passing headache, and even nasal burning. Topical corticosteroids such as Vancenase are effective in the treatment of rhinitis, especially for symptoms of itching, sneezing, and runny nose, but are less effective against nasal obstruction. Depending on the preparation, however, nasal corticosteroid sprays can also cause irritation, itching, burning, or sneezing. Local bleeding and septal perforation can also sometimes occur, especially if the spray is not properly targeted. Topical spheroids are generally more effective than cromolyn sodium in the treatment of allergic rhinitis. Immunotherapy, although expensive and inconvenient, often provides benefits, especially for patients who experience side effects with other medications. The so-called blocking antibodies, and agents that alter the release of cellular histamine, eventually result in decreased IgE, along with other favorable physiological changes. This effect is useful in IgE-mediated diseases, e.g., hypersensitivity in atopic patients with recurrent middle ear infections. Pulmonary fibrosis, interstitial lung disease (ILD), or interstitial pulmonary fibrosis, includes more than 130 chronic lung disorders that affect the lung, damaging lung tissue, and causing inflammation in the walls of air sacs in the lung., scarring or fibrosis in the interstitium (or tissue between the air sacs), and stiffness of the lung. Lack of breathing during exercise can be one of the first symptoms of these diseases, and a dry cough may be present. Neither the symptoms nor the X-rays are often sufficient to differentiate the different types of pulmonary fibrosis. Some patients with pulmonary fibrosis have known causes and some have unknown or idiopathic causes. The course of this disease is usually unpredictable and the disease is inevitably fatal. Their progress includes thickening and hardening of lung tissue, inflammation and difficulty in breathing. Most people may need oxygen therapy and the only treatment is lung transplantation.
Lung cancer is the most common cancer in the world. During 2003, there will be approximately 171,900 new cases of lung cancer (91,800 among men and 80,100 among women) in the United States alone and approximately 375,000 cases in Europe. Lung cancer is the leading cause of cancer death among men and women. There will be an estimated 157,200 deaths from lung cancer (88,400 among men and 68,800 among women) in 2003, accounting for 28% of total cancer deaths in the United States alone. More people die from lung cancer than from colon, breast and prostate cancers combined (American Cancer Society Web site, 2003, Detailed Guide: Lung Cancer: What are the Key Statistics?). Smoking tobacco is well established as the leading cause of lung cancer and it is believed that approximately 90% of cases are related to tobacco. There is a clear dose-response relationship between the risk of lung cancer and the number of cigarettes smoked per day, degree of inhalation, and age at which smoking began. Lifelong smokers have a 20-30 times higher risk of lung cancer than a non-smoker. However, the risk of lung cancer decreases over time after smoking is stopped. The relative risk of male ex-smokers decreases strongly with time from the end of exposure, but does not reach the risk of non-smokers, and does not decrease as much as for ex-smokers (Tyczynski et al., Lancet Oncol 4 (1): 45-55 (2003) Frequently, COPD and lung cancer are co-morbid diseases and the degree of underlying COPD can dictate whether a particular patient in a candidate for surgery. non-small cell lung cancer), only surgery (with or without radiation therapy or adjuvant chemotherapy) is curative. · the 1-year survival rate (the number of people who live at least 1 year after they were diagnosed with cancer) for lung cancer was 42% in 1998, largely due to improvements in surgical techniques • The 5-year survival rate for all stages of combined non-small cell lung cancer is of only 15% For small cell lung cancer, the relative survival rate in 5 years is approximately 6%. • For people who were found NSCLC and treated early with surgery, before it has spread to lymph nodes or other organs, the average 5-year survival rate is approximately 50%. However, only 15% of people with lung cancer are diagnosed at this early, localized stage. Obviously, there is much to improve in chemoprophylaxis. of lung cancer as well as the treatment of lung cancer. Dehydroepiandrosterone (DHEA) (3p-hydroxyandrost-5 ~ en-17-one) is a spheroid of natural origin secreted by the cortex. adrenal with apparent chemoprotective properties. Epidemiological studies have shown that low levels of endogenous DHEA correlate with the increased risk of developing some forms of cancer, such as premenopausal breast cancer in women and bladder cancer in both sexes. The ability of DHEA and DHEA analogs, such as DHEA-S (DHEA sulfate), to inhibit carcinogenesis is believed to result from its non-competitive inhibition of the enzyme activity of glucose-6-phosphate dehydrogenase (G6PDH). G6PDH is the rate-limiting enzyme of the hexose monophosphate pathway, a major source of intracellular ribose-5-phosphate and NADPH. Ribose-5-phosphate is a necessary substrate for the synthesis of both ribo- and deoxyribonucleotides. NADPH is a cofactor also involved in nucleic acid biosynthesis and the synthesis of hydroxymethylglutaryl Coenzyme A reductase (HMG CoA reductase). HG CoA reductase- is an unusual enzyme that requires two moles of NADPH per mole of product, the mevalonate, produced. Thus, it seems that the HMG CoA reductase would be ultrasensitive to the depletion of NADPH mediated by DHEA, and the cells treated with that DHEA would rapidly show the depletion of intracellular mevalonate wells. Mevalonate is required for DNA synthesis, and DHEA stops human cells in the Gl phase of the cell cycle in a manner closely resembling that of direct HMG CoA. Because G6PDH is required to produce mevalonic acid used in cellular processes such as protein isoprenylation and dolichol synthesis, a precursor to glycoprotein biosynthesis, DHEA inhibits carcinogenesis by depleting mevalonic acid and thus inhibits protein isoprenylation and glycoprotein synthesis. Mevalonate is the central precursor for cholesterol synthesis, as well as for the synthesis of a variety of compounds other than sterol involved in the post-translational modification of proteins such as farnesyl pyrophosphate and geranyl pyrophosphate; and for dolichol, which is required for the synthesis of glycoproteins involved in cell-to-cell communication and cell structure. It has been known for a long time that patients receiving steroid hormones of adrenocortical origin at pharmacologically appropriate doses show an increased incidence of infectious diseases. The U.S. Patent No. 5,527,789 describes a method for combating cancer by administering to a patient DHEA and ubiquinone, wherein the cancer is one that is sensitive to DHEA. DHEA is a 17-ketosteroid which is quantitatively one of the major adrenocorticoid steroid hormones found in mammals. Although DHEA seems to serve as an intermediate in the synthesis of gonadal steroids, the primary physiological function of DHEA has not been fully understood.
It is known, however, that the levels of this hormone begin to decline in the second decade of life (reaching 5% of the original level in old age.) Clinically, DHEA has been used systemically and / or topically to treat patients who suffer from psoriasis, gout, hyperlipidemia, and has been administered to post-coronary patients. In mammals, DHEA has been shown to have anticarcinogenic and weight-optimizing effects, and has been used clinically in Europe along with estrogen as an agent that reverses the symptoms of menopause and has also been used in the treatment of manic depression, schizophrenia, and Alzheimer's disease. DHEA has been used clinically at 40 mg / kg / day in the treatment of advanced cancer and multiple sclerosis. Moderate androgenic effects, hirsutism, and increased libido were the observed side effects. These side effects can be overcome by checking the dose and / or using analogues. The oral or subcutaneous administration of DHEA to improve the host response to infections is known, as is the use of the patch to deliver DHEA. DHEA is also known as a precursor to the metabolic pathway which ultimately leads to more powerful agents that increase the immune response in mammals. That is, DHEA acts as an immuno-modulator when it is converted to androstenediol or androst-5-en-3p, 17ß-γ1 (ß ???), and androstenetriol or androst-5-en-3p, 7β , 17p-triol (ß ???). However, DHEA in vitro has certain lymphoxic and suppressive effects on cell proliferation before its conversion to ß ??? and / or ß ???. Therefore, it is believed that the higher immunity enhancing properties obtained by administration of DHEA result from its conversion to more active metabolites. Adenosine is a purine involved in intermediary metabolism, and can be an important mediator in the lung for several diseases, including bronchial asthma, COPD, CF, RDS, rhinitis, pulmonary fibrosis, and others. The potential role of its receptor was suggested by the finding that asthmatics respond to atomized adenosine with marked bronchoconstriction while normal individuals do not. An asthmatic rabbit animal model, the asthmatic rabbit model allergic to the acaric acid powder for human asthma, responded in a similar manner to atomized adenosine with marked bronchoconstriction whereas non-asthmatic rabbits showed no response. A more recent work with this animal model suggested that adenosine-induced bronchoconstriction and bronchial hypersensitivity in asthma can be mediated primarily through the stimulation of adenosine receptors. Adenosine has also been shown to cause adverse effects, including death, when administered therapeutically for other diseases and conditions in subjects with hyperreactive airways not previously diagnosed. Adenosine plays a unique role in the body as a regulator of cellular metabolism. It can raise the cellular level of AMP, ADP and ATP that are energy intermediaries of the cell. Adenosine can stimulate or sub-regulate the activity of adenylate cyclase and thus cAMP levels. AMPc, in turn, plays an important role in the release of the neurotransmitter, cell division and the release of hormones. The main role of adenosine seems to be to act as a protective autocoid of lesions. In any condition in which ischemia, low oxygen tension or trauma occur, adenosine seems to play a role. It has been postulated that defects in the synthesis, release and / or degradation of adenosine contribute to the over activity of the amino acid neurotransmitters that excite the brain, and therefore to different pathological states. Adenosine has also been implicated as a primary determinant underlying the symptoms of bronchial asthma and other respiratory diseases, the induction of bronchoconstriction and the contraction of the smooth muscle of the airways. In addition, adenosine causes bronchoconstriction in asthmatics, but not in non-asthmatics. Other data suggest the possibility that adenosine receptors may also be involved in inflammatory and allergic responses by reducing the hyperactivity of the central dopaminergic system. It has been postulated that the modulation of signal transduction on the surface of inflammatory cells influences acute inflammation. It is said that adenosine inhibits the production of super oxide by stimulated neutrophils. Recent evidence suggests that adenosine may also have a protective role in stroke, CNS trauma, epilepsy, ischemic heart disease, coronary bypass, exposure to radiation, and inflammation. In general, adenosine seems to regulate cellular metabolism through ATP, act as a carrier for methionine, decrease the cellular oxygen demand and protect cells from ischemic injury. Adenosine is a tissue hormone or an intercellular messenger that is released when cells undergo ischemia, hypoxia, cell stress, and increased workload, and / or when the demand for ATP exceeds its supply. Adenosine is a purine and its formation is directly linked to the catabolism of ATP. It seems to modulate an arrangement of physiological processes that include vascular tone, hormonal action, neural function, platelet aggregation, and lymphocyte differentiation.
It also plays a role in the formation of DNA, the biosynthesis of ATP and the general intermediary metabolism. It is suggested that it regulates the formation of cAMP in the brain and in a variety of peripheral tissues. Adenosine regulates the formation of cAMP through two receptors i and A2. Adenosine reduces adenylate cyclase activity via receptors, although it stimulates adenylate cyclase in A2 receptors. The Ai receptors of adenosine are more sensitive to adenosine than the receptores receptors. It is generally believed that the CNS effects of adenosine are mediated by the Ai receptor, where peripheral effects such as hypotension, bradycardia, are said to be mediated by the A2 receptor. Few medications have been used for the treatment of respiratory diseases and conditions, although in general they all have limitations. Among them are glucocorticoid steroids, leukotriene inhibitors, anticholinergic agents, antihistamines, oxygen therapy, theophyllines, and mucolytics. Glucocorticoid steroids are the only ones with the widest use despite their well-documented side effects. Most of the available drugs, however, are effective in a small number of cases, and they are not effective for the treatment of asthma. There are no treatments currently available for many of the other respiratory diseases. Theophylline, an important drug in the treatment of asthma, is a known adenosine receptor antagonist which has been reported to eliminate adenosine-mediated bronchoconstriction in asthmatic rabbits. It has also been reported that a selective adenosine Al receptor antagonist, 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) inhibits adenosine-mediated bronchoconstriction and bronchial hypersensitivity in allergic rabbits. The therapeutic and preventive applications of the Al-specific adenosine receptor antagonists currently available are, however, limited by their toxicity. Theophylline, for example, has been widely used in the treatment of asthma, but it is associated with significant, frequent toxicity (gastrointestinal, cardiovascular, neurological and biological alterations) resulting from its narrow range of therapeutic doses. DPCPX far from being too toxic is clinically useful. The fact is that, despite decades of extensive research, no specific adenosine receptor antagonist is available for clinical use that certifies the general toxicity of these agents. Bronchodilators relax the muscle bands that wrap around the airways, allowing more air to enter and exit the lungs and improve breathing. Bronchodilators also help to clean the mucous membranes of the lungs and when the airways open, the mucous membranes move more freely and can be coughed or expelled more easily. There are forms of short-acting and long-acting bronchodilators; The short form of action mitigates or stops asthma symptoms while the long-acting form provides control of asthma symptoms and prevents asthma attacks. Bronchodilators include p2-agonists (short and long-acting forms), anticholinergics and theophyllines. Two commercially available anticholinergic bronchodilators are ipratropium bromide and tiotropium bromide. Ipratropium bromide is a quaternary ammonium compound with anticholinergic (parasympatholytic) properties. It seems to inhibit reflexes vaguely mediated by antagonizing the action of acetylcholine, a transmitting agent released from the vagus nerve. Anticholinergics prevent the increase in the intracellular concentration of cyclic guanosine monophosphate (G P cyclic) caused by the interaction of acetylcholine with the muscarinic receptor or the bronchial smooth muscle. Ipratropium bromide is commercially available as: Atrovent® (Boehringer Ingelheim) and Combivent® (Boehringer Ingelheim), in combination with albuterol sulfate.
Tiotropium bromide is commercially available as Spiriva® (Boehringer Ingelheim and Pfizer). Spiriva® is a once-daily inhaled COPD treatment that works by prolonged blocking of the M3 receptor. Spiriva® was launched in June 2002 in five countries in Europe (Denmark, Finland, Germany, Holland and Sweden) and by the end of the year had been launched in 13 countries around the world. The combined data from two six-month studies showed that Spiriva® was statistically superior to Salmaterol for average and peak LVEF (forced expiratory volume in one second) and FVC (forced vital capacity) (Boehringer Ingelheim, May 5, 2003 releases pressure ). The U.S. Patent No. 5, 660,835 (and the corresponding PCT publication WO 96/25935) discloses a new method for treating asthma or adenosine depletion in a subject by administering to the subject a dehydroepiandrosterone (DHEA) or a compound related to DHEA. The patent also discloses a pharmaceutical composition in reference to an inhalable or breathable formulation comprising DHEA or DHEA-related compounds that is in a breathable particle size .. U.S. Pat. No. 5,527,789 discloses a method of combating cancer in a subject by administering to a subject a DHEA or a compound related to DHEA, and ubiquinone to combat heart failure induced by DHEA or a compound related to DHEA. The U.S. Patent No. 6,087,351 describes an in vivo method for the reduction or depletion of adenosine from the tissue of a subject by administering to the subject a DHEA or a compound related to DHEA. The U.S. patent application Series No. 10 / 454,061, filed June 3, 2003, describes a method for treating COPD in a subject by administering to the subject a DHEA or a compound related to DHEA. The U.S. patent application Series No. 10 / 462,901, filed June 17, 2003, discloses a stable dry powder formulation of DHEA in a nebulizable form sealed in a container. The U.S. patent application Series No. 10 / 462,927, filed June 17, 2003, discloses a stable dry powder formulation of DHEA-S, in the form of a dihydrated crystal, suitable for treating asthma and COPD. The above patents and patent applications are hereby incorporated by reference in their entirety. There is a well-defined need for effective and novel therapies to treat respiratory ailments, lung and cancer that to date can not be treated, or at least for which there are no therapies available that are effective and that are free of significant harmful side effects. . This is the case of insufficiencies or ailments that affect the respiratory system, and more particularly the lung and the pulmonary tract, which include breathing difficulties, asthma, bronchoconstriction, pulmonary inflammation, and allergies, depletion or hyposecretion of the surfactant, etc. in addition, there is a definite need for treatments that have prophylactic and therapeutic applications, and require low amounts of active agents, which makes them less expensive and less prone to harmful side effects. In addition, there is a need to ensure that the patient complies with taking their medication, and a need to facilitate the taking of the plurality of compounds necessary to prevent or treat asthma, COPD or other respiratory diseases. SUMMARY OF THE INVENTION The present invention provides a composition comprising at least two active agents. A first active agent comprises a non-glucocorticoid spheroid, such as an epiandrosterone (EA) or a salt thereof. A second active agent comprises an anticholinergic bronchodilator. The composition comprises a combination of the first active agent and the second active agent. The amount of the first active agent and the amount of the second active agent in the composition is of an amount sufficient to effectively or prophylactically treat a subject in danger of suffering or suffering from asthma., COPD, or other respiratory diseases when the composition is administered to the subject. The composition may further comprise other bioactive ts and formulation ingredients. The composition is a pharmaceutical or veterinary composition suitable for administration to a subject or patient, such as a human or a non-human animal (such as a non-human mammal). The composition is useful for treating asthma, COPD, or other respiratory diseases for which inflammation and its sequelae play a role that includes conditions associated with broncho-constriction, surfactant depletion and / or allergies. The present invention also provides methods for treating asthma, COPD, lung cancer, or other respiratory diseases comprising administering the composition to a subject in need of such treatment. The present invention also provides a use of the first active t and the second active t in the manufacture of a medicament for the prophylactic or therapeutic treatment of asthma, COPD or other respiratory diseases described above. The present invention also provides a kit comprising the composition and a device for delivering it. The delivery device is capable of delivering the composition to the subject. Preferably, the delivery device comprises an inhaler provided with an aerosol or spray-generating medium that supplies the particles. Preferably, the supply is to the subject's airways. More preferably, the supply is for the lung or the lungs of the subject. Preferably, the supply is directly to the desired location. The main advantof using the composition is the compliance of patients in need of such prophylaxis or treatment. Respiratory diseases such as asthma or COPD are multifactorial with different manifestations of signs and symptoms for individual patients. As such, most patients are treated with multiple medications to alleviate different aspects of the disease. A fixed combination of the first active t, such as DHEA or DHEA-S, and the second active t, such as ipratropium or tiotropium, allows a more convenient target therapy for a defined patient subpopulation. Patient compliance should be improved by simplifying and focusing on each patient's unique disease attributes such that their specific symptoms are addressed in the most expeditious manner. In addition, there is an added advantof convenience or time saving in the administration of both, the first and second active ts in an administration. This is specifically true when the composition is administered to a region of the subject's body that has the potential for discomfort, such as the composition administered to the subject's airways. This is also especially true when the administration of the compositions to the subject is aggressive. In addition, the first active t, such as DHEA or DHEA-S, is an anti-inflammatory t that is more effective when it is delivered or deposited in the distal peripheral airways rather than in the airway conduction, in the alveolar membranes and in the fine airways. Patients with asthma and some with COPD have air conduction pass that are shrunk, which limits the supply (due to the previous deposition caused by a lower particle velocity) of the first active t, such as DHEA, which acts in these distal peripheral airways. Therefore, the combination of a bronchodilator drug (p2-agonist, antimuscarinic which invests a high tone) facilitates the delivery of an anti-inflammatory to the distal peripheral airways. The use of the combination provides an improved sustained pharmacological effect that results in improved disease control. Antileukotrienes reduce interstitial edema in very small peripheral airways. This would also have the effect of increasing the diameter of the peripheral airways and facilitating the delivery of the first active t. This is also true for antihistamines, which also reduce the edema of the peripheral airways and facilitate the supply of the first active agent to the distal airways. The drawings accompanying this patent form part of the description of the invention, and further illustrate some aspects of the present invention as discussed below. BRIEF DESCRIPTION OF THE FIGURES Figure 1 represents the fraction of fine particles of DHEA-S "2H20 micronized pure supplied from a single-dose Acu-Breathe inhaler, as a function of the flow rate.The results are expressed as DHEA-S The IDL data in the virtually anhydrous micronized DHEA-S are also shown in this figure where the result of 30 L / min was set to zero since no detectable mass enters the impactor Figure 2 represents the chromatograms HPLC of DHEA-S bulk virtually anhydrous after being stored pure and mixed with lactose for 1 week at 50 ° C. Control was pure DHEA-S stored at room temperature (TA) Figure 3 represents HPLC chromatograms for DHEA -A.'2H2 <0 in bulk after being stored pure and mixed with lactose for 1 week at 50 ° C. The control was pure DHEA-A "2H2Ü stored at room temperature (TA) Figure 4 depicts the solubility of DHEA-S as a function of NaCl concentration at two temperatures Figure 5 depicts the solubility of DHEA-S as a function of the concentration of the reciprocal sodium cation at 24-25 ° C. Figure 6 represents the solubility of DHEA-S as a function of the concentration of the sodium cation reciprocating at 7-8 ° C. Figure 7 represents the solubility of DHEA-S as a function of the concentration of NaCl with and without buffer at RT Figure 8 represents the solubility of DHEA-S as a function of the concentration of the sodium cation reciprocating at 24-25 ° C with and without Figure 9 represents the concentration of the DHEA-S solution versus time under two storage conditions Figure 10 represents the concentration of the DHEA solution versus time under two storage conditions.
Figure 11 depicts a scheme of nebulization experiments. Figure 12 represents the mass of DHEA-s deposited in a bypass manifold as a function of the initial concentration of the solution placed in the nebulizer. Figure 13 represents the particle size by cascade impaction for the solutions of the DHEA-S nebulizer. The presented data are the average of the 7 nebulization experiments. Figure 14 depicts the inhibition of HT-29 SF cells by DHEA. Figure 15 depicts the effects of DHEA on the cell cycle distribution in HT-29 SF cells. Figures 16a and 16b depict the reversal of DHEA-induced growth inhibition in HT-29 cells. Figure 17 represents the inversion of Gl induced by interrupted DHEA in the HT-29 SF cells. Figure 18 depicts the effect of anticholinergic agents and DHEA-S on ASM cells. Figure 19 represents certain suitable analogs of DHE. Figure 20 depicts certain suitable analogs of DHEA.
Figure 21 represents certain suitable analogs of DHEA. Figure 22 represents suitable modifications of the ketone C-17 of DHEA. DETAILED DESCRIPTION OF THE INVENTION Definiclons In the present context, the terms "adenosine" and "surfactant" depletion have the purpose of encompassing the levels that decrease or deplete in the subject compared with the previous levels in that subject, and the levels that they are essentially the same as the previous levels in that subject but, due to some other reason, a therapeutic benefit would be achieved in the patient by modifying the levels of these agents compared to the previous levels. The term "airway", as used herein, means that part or all of a subject's respiratory system is exposed to air. The term airway includes, but not exclusively, trachea, tracheobronchial tree, nasal passages, sinuses, among others. The airway also includes trachea, bronchi, bronchioles, terminal bronchioles, respiratory bronchioles, alveolar ducts and alveolar sacs. The term "airway inflammation" as used herein, refers to a disease or condition related to inflammation in the subject's airways. Inflammation of the airways may be caused or be accompanied by allergy (s), asthma, impaired respiration, cystic fibrosis (CF), chronic obstructive pulmonary disease (COPD), allergic rhinitis (AR), acute respiratory distress syndrome (ARDS). ), viral or microbial infections, pulmonary hypertension, lung inflammation, bronchitis, cancer, obstruction of the airways and bronchoconstriction. The term "carrier", as used herein, means a biologically acceptable carrier in the form of solid, liquid, gaseous carriers, and mixtures thereof, which are suitable for the various proposed routes of administration. Preferably, the carrier is pharmaceutically or veterinarily acceptable. "An effective amount" as used herein means an amount that provides a therapeutic or prophylactic benefit. "Other therapeutic agents" refer to any therapeutic agent that is not the first or the second active agent of the composition. The term "prophylaxis," as used herein, means a prophylactic treatment effected before a subject experiences a disease or worsening of a previously diagnosed condition such that a subject can avoid, prevent or reduce the likelihood of having a symptom of the disease. or condition related to it. The subject has the increased risk of obtaining the disease or a worsening of a previously diagnosed condition. The term "respiratory disease", as used herein, means diseases or conditions related to the respiratory system. Examples include, but are not limited to, airway inflammation, allergy (s), impaired breathing, cystic fibrosis (CF), allergic rhinitis (AR), acute respiratory distress syndrome (ARDS), cancer, pulmonary hypertension, lung inflammation, bronchitis, obstruction of the airways, bronchoconstriction, microbial infection and viral infection such as MRS . The term "treat", "treatment" or "therapeutic", as used herein, means a treatment that decreases the likelihood that the subject to whom such treatment was administered will manifest symptoms of disease or other condition. The present invention provides a composition comprising a first active agent comprising a non-glucocorticoid steroid, such as an epiandrosterone (EA), analog thereof, or a salt thereof (preferably DHEA or DHEA-S), in combination with a second active agent comprising an anticholinergic bronchodilator. The composition may further comprise a pharmaceutically or veterinarily acceptable carrier, diluent, excipient, agent or bioactive ingredient. The compositions are useful for the treatment of asthma, COPD, or other respiratory diseases. Other respiratory diseases for which treatment the compositions are also useful, are respiratory and pulmonary diseases and conditions associated with bronchoconstriction, lung inflammation and / or allergies, and lung cancer. The first active agent is an epiandrosterone, an analog or a pharmaceutically or veterinarily acceptable salt thereof. The epiandrosterone, an analog or a pharmaceutically or veterinarily acceptable salt thereof is selected from a non-glucocorticoid spheroid having the chemical formula where the broken line represents a single or double bond; R is hydrogen or a halogen; H is position 5 is present in the alpha or beta configuration or the compound of chemical formula I comprises a racemic mixture of both configurations; and ¾ is hydrogen or a multivalent organic or inorganic dicarboxylic acid covalently linked to the compound; a non-glucocorticoid steroid of the chemical formula: a non-glucocorticoid steroid of the chemical formula: a combination thereof, wherein Ri, R2, R3, 4, R5 / 7, s, R9, io, R12, R13, 1 and R19 are independently H, OR, halogen, Ci_10 alkyl or Ci-10r alkoxy R5 and R12 are independently OH, SH, H, halogen, pharmaceutically acceptable ester, pharmaceutically acceptable thioester, pharmaceutically acceptable ether, pharmaceutically acceptable thioether, pharmaceutically acceptable inorganic esters, pharmaceutically acceptable monosaccharide, disaccharide or oligosaccharide, spiro-oxirane, spirothirane, -OSO2R20, -OPOR20R21 or Ci_io alkyl, R5 and R6 taken together are = 0, Rio and Rn taken together are = 0; R15 is (1) H, halogen, C1-C10 alkyl, or Ci-C10 alkoxy when Ras is -C (0) 0R22, (2) H, halogen, OH or C1-C10 alkyl when Ri6 is halogen, OH, or Ci-Cio alkyl, (3) H, halogen, Ci-C10 alkyl, C-alkenyl, C1-C10 alkynyl, formyl, Ca-Cio alkanoyl or epoxy when Ri6 is OH, (4 ) OR, SH, H, halogen, pharmaceutically acceptable ester, pharmaceutically acceptable thioester, pharmaceutically acceptable ether, pharmaceutically acceptable thioether, pharmaceutically acceptable inorganic esters, monosaccharide, pharmaceutically acceptable disaccharide or oligosaccharide, spiro-oxirane, spirothirane, -OSO2R20 or -OPOR20R21 when Ri6 is H, or R15 and Ra6 taken together are = 0; R17 and Rae are independently (1) H, -OH, halogen, Ci-C alkyl or or Ca-10 alkoxy when R6 is H, OR, halogen, Ci-C10 alkyl or -C (0) OR22 (2) H, (Ca-Cao alkyl) · amino, (C-C o) namino- alkyl (C-o-alkyl), C-C-alkoxy, hydroxy-C2-C alkyl, alkoxy of Ca-Cio-Ci-C10 alkyl, (halogen) mal of Ca-Cao / - alkanoyl of Ca-Cao formyl, carbalkoxy of C -Co or Ci-Co alkanoyloxy when Rs and R 6 taken together are = 0, (3) R17 and Rs taken together are = 0; (4) Ra7 or Ras taken together with the carbon to which they are attached form a 3 to 6 member ring containing 0 or 1 oxygen atom; or (5) Rs and Ra7 taken together with the carbons to which they are attached form an epoxide ring; R20 and R21 are independently OH, pharmaceutically acceptable ester or pharmaceutically acceptable ether; R 22 is H, (halogen) m- (Ca-a alkyl) or Ca-10 alkyl; n is 0, 1 6 2; and m is 1, 2 or 3; or pharmaceutically or veterinarily acceptable salts thereof. Preferably, for the chemical formula (I), the multivalent organic dicarboxylic acid is SO2OM, phosphate or carbonate, wherein M comprises a counterion. Examples of a counterion are H, sodium, potassium, magnesium, aluminum, zinc, calcium, lithium ammonium, amine, arginine, lysine, histidine, triethylamine, ethanolamine, choline, triethanolamine, procaine, benzathine, tromethanin, pyrrolidine, piperazine, diethylamine, sulfatide and phosphatide -P-OC¾CHCH2OCÓR% «I O OCOR2 where R2 and. R3, which may be the same or different, are linear or branched? -0-O4 alkyl or glucuronide The hydrogen atom at the 5-position of the chemical formula I may be present in the alpha or beta configuration, or the DHE¾ compound may be provided as a mixture of compounds of both configurations. Illustrative compounds of the chemical formula I above include, but are not limited to, DHEA, wherein R and R1 are each hydrogen, containing a double bond; 16-alpha bromoepiandrosterone, wherein R is Br, R 1 is H, which contains a double bond; 16-alpha-fluoro epiandrosterone, where R is F, R1 is H, which contains a double bond; etiocolanolone, wherein R and R1 are each hydrogen that lacks a double bond; and dehydroepiandrosterone sulfate, wherein R is H, R1 is S02OM and M is a sulfatide group as defined above, which lacks a double bond. Others, however, are also included. The also preferred compounds of formula I are those wherein R is halogen, e.g. bromine, chlorine or fluoro, where R1 is hydrogen, and where a double bond is present. The most preferred compound of formula I is 16-alpha-fluoro epiandrosterone. Other preferred compounds are DHEA and salts of DHEA, such as the sulfate salt (DHEA-S). In general, the non-glucocorticoid steroid, such as those of the formulas (I), (III) and (IV), their derivatives and their salts are administered in a dose of about 0.05, about 0.1, about 1, about 5, about 20 to about 100, about 500, about 1000, about 1500, about 1800, about 2500, about 3000, about 3600 mg / kg of body weight. Other doses, however, are also suitable and contemplated within this patent. The first active agent of formula (I), (III) and (IV) can be prepared according to known procedures, or variations thereof which will be apparent to those skilled in the art. See, for example, U.S. Pat. No. 4,956,355; UK Patent No. 2,240,472; EPO Patent Application No. 429; 187, PCT Patent Publication No. WO 91/04030; U.S. Patent No. 5,859,000; Abou-Gharbia et al., J. Pharm. Sci.70: 1154-1157 (1981); Merck Index onograph No. 7710 (11th Ed. 1989), among others. In some embodiments of the invention, the first active agent may be an epiandrosterone analog or derivative thereof. Also, prodrugs and active metabolites of epiandrosterone are included in the present invention. Those skilled in the art will recognize that the compounds described herein can show the phenomena of tautomerism, conformational isomerism, geometric isomerism and / or optical isomerism. It should be understood that the invention encompasses any of the tautomeric, isomeric, conformational, isomeric, and / or geometric isomeric forms of the compounds having one or more of the utilities described herein, as well as mixtures of these different forms.
The metabolites of epiandrosterones, such as those described in the following references, can be used as the first active agent - Capillary gas chromatography of urinary steroids ofterbutaline-treated asthmatic children, Chromatographia (1998), 48 (1/2), 163-165; Androstenedione metabolism in human lung fibroblasts; Journal of Steroid Biochemistry (1986), 24 (4), 893-7; Metabolism of androsterone and 5a-androstane-3a, 1 (3-diol in human lung tissue and in pulmonary endothelial cells in culture, Journal of Clinical Endocrinology and Metabolism (1985), 60 (2), 244-50; Testosterone metabolism by human lung tissue, Journal of Steroid Biochemistry (1978), 9 (1), 29-32; Metabolism of androsterone and 5 alpha-androstane-3 alpha, 17 beta-diol in human lung tissue and in pulmonary endothelial cells in culture, Journal of clinical endocrinology and metabolism (1985 Feb), 60 (2), · 44-50; Metabolism of dehydroisoandrosterone and androstenedione in human pulmonary endothelial cells in culture, Journal of clinical endocrinology and metabolism (1983 May), 56 (5), 930- 5; Metabolism of dehydroisoandrosterone and androstenedione by the human lung in vitro, Journal of steroid biochemistry (1977 Apr), 8 (4), 277-84; and Testosterone metabolism in dog lung in vitro, Steroids and lipids research (1973), 4 (1), 17-23, which are incorporated here in their totality d as a reference. Other suitable analogues of DHEA that can be used as the first active agent are described herein. Figure 19 depicts certain suitable analogs of DHEA, including the compounds of formulas IA, IB, IC, and ID. In the representation of the appropriate R groups, the point of attachment is indicated by a C¾ group or by an atom marked with an asterisk. Ri and R3 may be linear or branched alkyls including benzyl and optionally substituted alkyls, such as aminoalkyls, hydroxyalkyls, ethers and carboxylic acids, and optionally substituted aryl and heteroaryls. Ri and R3 can be, for example CF3 CH3 (CH2) n wherein n is preferably 0 to 4 wherein X = OH, O-alkyl, N (substituted by H, alkyl or acyl) and m and p are independently 1 to 4, wherein Y = O, NH, N -alkyl or N-acyl C02H or amide Examples of compounds of formula IA include, R2 is preferably a substituent derived from diacid or amino acid derivative, which potentially includes chloroacetyl derivatives and optionally substituted acrylate derivatives or aryls such as benzyl and heterobenzoyl. Examples of compounds of formula IB include Examples of compounds of the formula IC include, R4 can be aromatic in nature and examples of suitable compounds of formula ID include, Other suitable analogs include analogs where modifications have been made at the C-3 position by retaining OH or replacing OH with NH. These analogs are typically prepared starting from andrst-4-ene-3,17-dione protected with C17 acetal, as represented in Figure 20. The compounds of formula 1E can be derived from Grignard reagents and possibly aryl lithium reagents, such as aromatics by nature, and may also be alkynyl, alkenyl and alkyl. Examples of R5 are CH3 (CH2) n where n = 0 to 4 Examples of compounds of formula 1E include Rg and Rg can independently be a diverse group of amines and can include amines having the functionalities described for the Ri group. Examples of suitable compounds of formula 1F include, Suitable R7 groups can be derived from Grignard / organolithium reagents and thus could comprise the functionalities described for R5. Examples of compounds of formula 1G include Examples of compounds of formula 1H include Other suitable analogs include those compounds wherein the C-2 position of DHEA was modified. Suitable modifications are shown in Figure 21. R9 can be derived from alkylating agents, such as alkyl, benzyl, heterobenzyl and derivatives of other activated halides. Examples of R9 include, Examples of compounds of formula 1J include Rio can be aromatic esters such as with aryl or heteroaryl ring or enolisable alkyl esters. Examples of compounds of formula 1K include.
R n can be a series of aromatic and heteroaromatic aldehydes such as benzene carboxyaldehyde and substituted variants thereof, pyridine carboxyaldehyde or non-enolisable aldehydes such as (CH 3) 3 CCH = 0. Examples of compounds of formula IL include, R12 can be a subgroup of an amine such as for R6. Examples of compounds of formula 1N include, Suitable modifications of the C17 ketone of DHEA are depicted in Figure 22. The compounds depicted in Figure 22 can also be used as the first active agent. Other suitable analogs of DHEA are described in U.S. Pat. No. 6 / 635,629; European Patent 934745; Dehydroepiandrosterone and analogs inhibit DNA binding of AP-1 and airway smooth muscle proliferation, Journal of Pharmacology and Experimental Therapeutics (1998), 285 (2), 876-883; and Dehydroepiandrosterone and related steroids inhibit mitochondrial respiration in vitro, International Journal of Biochemistry (1989), 21 (10), 1103-7, which are incorporated herein in their entirety. The second active agent is an anticholinergic bronchodilator. The bronchodilator is an ipratropium or tiotropium. Preferably, ipratropium is an ipratropium halide. Preferably, the tiotropium is a tiotropium halide. The halide is chloride, bromide or iodide. More preferably, ipratropium is an ipratropium bromide. More preferably, tiotropium is a tiotropium bromide. The anticholinergic bronchodilator has the formula wherein: Ri is C2-C4 alkyl, C2-C4 alkenyl or C3-C8 cycloalkyl, preferably methyl, R2 is Ci-C4 alkyl, preferably methyl or isopropyl,? ¾ is hydrogen or acyl, preferably acetyl or benzoyl , preferably hydrogen, and X "the anion of a monovalent or polyvalent acid, preferably of a hydrohalic acid A preferred compound is ((8r) ~ 8-isopropyl-3a- [(+) - tropoyl-oxy] -IccH, H-tropanium) of ipratropium defined by the following formula: Preferably, ipratropium is ipratropium bromide (also known as N-isopropyl nortropintropic acid methobromide). Preferably, the daily dose of ipratropium is between 150 to 500 μg, preferably 200 to 350 μg. In the case of a metered aerosol which supplies, for example, 20 xg ipratropium bromide per drive, two or possibly three drives are atomized with a single dose of 40 to 60 g three to four times daily in each nostril, such that a total daily dose between 240 to 480 μg is achieved. The compounds of the chemical formula (V) are prepared and isolated by the method described in U.S. Pat. No. 3,505,337 and 4,385,048 (the description of which are incorporated herein by reference). Ipratropium bromide is commercially available under the tradename Atrovent® (Boehringer Ingel eim Pharmaceuticals, Inc., Ridgefield, CT). Bronchodilation after inhalation of Atrovent® is induced by local drug concentrations sufficient for an anticholinergic efficacy in the bronchial smooth muscle and not by systemic drug concentrations. In 90-day controlled studies in patients with bronchospasm associated with COPD (chronic bronchitis and emphysema) significant improvements in lung function (increases in LVEF and FEF25-75% of 15% or more) occurred in 15 minutes, reaching a peak in 1 - 2 hours, and persisted in most patients for up to 6 hours. In 90-day controlled studies in patients with bronchospasm associated with asthma, significant improvements in lung function occurred (increases in LVEF of 15% or more) in 40% of patients. Preclinical and clinical evidence suggest that there are no harmful effects of Atrovent® on the mucous secretion of airways, mucociliary clearance or gas exchange. The bronchodilator effect of Atrovent® in the treatment of acute bronchospasm associated with asthma has been demonstrated in studies in children older than 6 years. In most of these studies Atrovent® was administered in combination with an inhaled beta agonist. Although the data are limited, Atrovent® has shown to have a therapeutic effect in the treatment of bronchospasm associated with viral bronchiolitis and bronchopulmonary dysplasia in infants and very young children. Ipratropium bromide is also commercially available under the trade name Combivent® Aerosol Inhalation (Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT). The Combivent® inhalation spray also includes the p2-adrenergic bronchodilator, albuterol sue, (also known as salbutamol). The means of administration is by inhalation. The usual dose of the Combivent® inhalation spray is two inhalations four times a day, up to 12 puffs in 24 hours. Each dose contains approximately 18 μg of ipratropium bromide and 120 μg of albuterol sue. The anticholinergic bronchodilator also has the formula (VI): wherein: R1 is a thiophenyl, phenyl, furyl, cyclopentyl or cyclohexyl, or a thiophenyl or phenyl substituted with methyl, or thiophenyl or phenyl substituted with chloro or fluoro, R2 is a hydroxyl, Ci_4 alkyl or Ci_4 alkoxy / Ra is a hydrogen, fluoro, chloro, or methyl, Q is -CH2CH2-, -CH2CH2CH2-, -CH = CH-, or HH-C- \ /, Q 'is -NR- or -N + RR'-, wherein R is hydrogen, halogen or substituted Ci-4 alkyl or hydroxyl, wherein R' is a C1-4 alkyl, wherein the positive charge of the Nitrogen atom is coupled with an equivalent anion (X ~), where X "is an anion of a monovalent or polyvalent acid, preferably of a hydrohalic acid, and m is 1 or 2. Preferably, the thiophenyl is covalently linked to the chain of carbon at the a-thiophenyl position Preferably, R 1 is a thiophenyl.Preferably, R 2 is hydroxyl Preferably, R a is hydrogen Preferably Q is H H -C -C- / O Preferably, Q 'is -N + RR Preferably, R is methyl and R 'is methyl Preferably, X "is Br ~. Preferably, m is 1. A preferred compound is tiotropium ((1R, 2R, 4S 5S, 7S) -7- [2-hydroxy-2, 2-di (2-thienyl) acetoxy] -9,9-dimethyl- 3 oxa-9-azoniatricyclo [3.3.1. O2.4] onane or 6b, 7b-epoxy-3a- [2-hydroxy-2, 2-di (2-thienyl) acetoxy] -8,8-dimethyl-H, 5aH-tropanio) which is defined by the following formula Preferably, the tiotropium is tiotropium bromide. The compounds of the chemical formula (VI) are prepared and isolated by the method described in European Patent Application No. EP 0 418 716 Al (the description of which is incorporated herein by reference). The compound of the chemical formula (VIII) can also be prepared and isolated by the methods described in Inn, R., Drugs of the Future 25 (7): 693-699 (2000) (this article can be found at the electronic address: http: //www.prous. com / journals / dof / sample / tml / df250693 / df250693.html) (the description of which is incorporated here for reference). Tiotropium bromide is commercially available as Spiriva® (Boehringer Ingelheim and Pfizer). The preferred dose for tiotropium bromide is 10-160 μg. The preferred means of administration is by inhalation. The first and second active agents are used to treat respiratory and pulmonary diseases, and any of the additional agents listed below, may be administered per se or in the form of pharmaceutically acceptable salts, as discussed above, all referred to as "compounds or active agents ". The first and second active agents can also be administered in combination with each other, separately, or bound in, pharmaceutically or veterinarily acceptable formulation (s). The active compounds or their salts can be administered either systemically or topically, as discussed below. The present invention also provides methods for the treatment of asthma, COPD, or other respiratory diseases, comprising administering the composition to a subject in need of such treatment. The method is for prophylactic or therapeutic purposes. The method comprises an in vivo method. The method is effective to treat a plurality of diseases, whatever their cause, including administration of spheroids, abnormalities in the synthesis or metabolism of adenosine or the adenosine receptor, or any other cause. The method includes treating respiratory and lung diseases, either by reducing adenosine or adenosine receptor levels, by reducing hypersensitivity to adenosine, or any other mechanism, particularly in the lung, liver, heart and brain, or any organ that is in need of such treatment. Other respiratory diseases referred to herein include cystic fibrosis (CF), dyspnea, emphysema, asthmatic breathing, pulmonary hypertension, pulmonary fibrosis, lung cancer, hypersensitive airways, increased adenosine or adenosine receptor levels., particularly those associated with infectious diseases, pulmonary bronchoconstriction, lung inflammation, lung allergies, surfactant depletion, chronic bronchitis, bronchoconstriction, difficulty breathing, obstructed and impaired lung pathways, adenosine test for cardiac function, pulmonary vasoconstriction, impaired breathing, acute respiratory distress syndrome (ARDS), administration of certain drugs, such as adenosine and drugs that increase the level of adenosine and other drugs for eg treat supra ventricular tachycardia (SVT), and the administration of adenosine stress tests, infant respiratory distress syndrome (childhood SDR), pain, allergic rhinitis, decreased lung surfactant, severe acute respiratory syndrome (SARS), among others.
In one embodiment, the invention is a method for the prophylaxis or treatment of asthma which comprises administering the composition to a subject in need of such treatment an amount of the composition sufficient for the prophylaxis or treatment of asthma in the subject. In one embodiment, the invention is a method for the prophylaxis or treatment of COPD which comprises administering the composition to a subject in need of such treatment an amount of the composition sufficient for the prophylaxis or treatment of COPD in the subject. In one embodiment, the invention is a method for the prophylaxis or treatment of bronchoconstriction, pulmonary inflammation or pulmonary allergy which comprises administering the composition to a subject in need of such treatment an amount of the composition sufficient for the prophylaxis or treatment of bronchoconstriction, inflammation pulmonary or pulmonary allergy in the subject. In one embodiment, the invention is a method for the reduction or depletion of adenosine in the tissue of a subject which comprises administering the composition to a subject in need of such treatment an amount of the composition sufficient to reduce or deplete adenosine in the tissue of a subject. The present invention also provides a use of the first active agent and the second active agent in the manufacture of a medicament for the treatment of asthma, COPD or other respiratory diseases, including lung cancer. The medicament comprises the composition described through this description. The daily dose of the first active agent and the second active agent to be administered to a subject will vary with the total treatment schedule, the first active agent and the second active agent to be employed, the type of formulation, the route of administration and the condition of the patient. Examples 11 to 18 show aerosol preparations according to the invention for delivery with a device for nasal or respiratory administration or administration by inhalation. For intrapulmonary administration, liquid preparations are preferred. In the case of other bioactive agents, there are amounts recommended by the FDA to supplement the dietary intake of a person with additional bioactive agents, such as in the case of vitamins and minerals. However, where it is used for the treatment of specific conditions or to improve the immune response of a subject can be used in doses one hundred and one thousand times higher. Most of the recommendations of the pharmacopoeia cover a very wide range of doses, from which the doctor can draw a guide. The amounts for the exemplary agents described in this patent may be in the range of those commonly recommended for daily consumption, lower or higher than those levels. Treatment can typically begin with a low dose of a bronchodilator in combination with a non-glucocorticoid steroid, or other bioactive agents as appropriate, and then a dosage assessment for each patient. However, larger and smaller amounts, including initial amounts, may also be administered, also within the limits of this invention. The preferred ranges for the first and second active agents, or any other therapeutic agent, employed herein will vary depending on the route of administration and the type of formulation employed, as judged by the physician and the manufacture will be in accordance with known methods and components. The active compounds can be administered as a dose (once a day) or in several doses (several times a day). The compositions and method for preventing and treating respiratory, cardiac and cardiovascular diseases can be used to treat adults and infants, as well as non-human animals affected with the conditions described. Although the present invention is mainly concerned with the treatment of human subjects, it can also be used for veterinary purposes in the treatment of non-human mammalian subjects, such as dogs and cats as well as for large wild and domestic animals. The terms "high" and "low" levels of "adenosine" and "adenosine receptors" as well as "adenosine depletion" are intended to include both conditions where adenosine levels are higher than, or lower (still depleted) when compared to previous adenosine levels in the same subject, such as conditions where adenosine levels are within the normal range but, due to some other condition or alteration in that patient, a therapeutic benefit would be obtained in the patient decrease or increase adenosine or adenosine receptor levels or hypersensitivity. Thus, this treatment helps to regulate (titrate) the patient in a custom-made way. Although administration of the first active agent can also decrease or reduce adenosine levels in a subject having either normal or higher levels prior to treatment, additional administration of the second active agent will improve the patient's breathing in a short period of time. The further addition of other therapeutic agents will help to titrate or titrate the undesirably low levels of adenosine, which can be observed with the administration of the present treatment, particularly until reaching the optimal titration of the appropriate dose.
Other therapeutic agents that may be incorporated within the present composition are one or more of a variety of therapeutic agents that are administered to humans and animals. The composition may comprise, in addition to the first and second active agents, a ubiquinone and / or folic acid. A ubiquinone is a compound represented by the formula: or a pharmaceutically acceptable salt thereof. Preferably, the ubiquinone is a compound according to the chemical formula provided above, wherein n = l-10 (Coenzymes Qi-io) / more preferably n = 6-10, (Coenzymes Qs- ??) and more preferably n = 10 (Coenzyme Q10) · Ubiquinone is administered in a therapeutic amount to treat the disease or target condition, and the dosage will vary depending on the condition of the subject, other agents that are administered, type of formulation, and the route of administration. Ubiquinone is preferably administered in a total amount per day of about 0.1, about 1, about 3, about 5, about 10, about 15, about 30 to about 50, about 100, about 150, about 300, about 600, about 900 , approximately 1200 mg / kg of body weight. More preferably the total amount per day is from about 1 to about 150 mg / kg, about 30 to about 100 mg / kg, and more preferably about 5 to about 50 mg / kg. Ubiquinone is a natural substance and is commercially available. The active agents of this invention are provided within wide ranges of amounts of the composition. For example, the active agent may be contained in the composition in amounts of about 0.001%, about 1%, about 2%, about 5%, about 10%, about 20%, about 40%, about 90%, about 98% , approximately 99.999% of the composition. The amount of each active agent can be adjusted when, and if, additional agents with activities that overlap as described in this patent are included. The dosage of the active compounds, however, can vary depending on the age, weight and condition of the subject. The treatment can be started with a small dose, e.g. less than the optimum dose, of the first active agent of the invention. This can be effected in a similar manner with the second active agent, until reaching the desired level. Or vice versa, for example in the case of multivitamins and / or minerals, the subject can be stabilized at a desired level of these products and subsequently administer the first active compound. The dose can be increased until an optimum and / or desired effect is reached according to the circumstances. In general, the active agent is preferably administered in a concentration that will provide effective results without causing any side effects that are unduly dangerous or harmful, and can be administered either as a single unit dose, or if desired in convenient subunits administered at times appropriate to 'throughout the day. The second therapeutic or diagnostic agent (s) is (are) administered in amounts known in the art to be effective for the intended application. In cases where the second active agent has an activity that overlaps with the principal agent, the dose of one of the other or both agents can be adjusted to obtain the desirable effect without exceeding a dose range that avoids adverse side effects. Thus, for example, when other analgesic and anti-inflammatory agents are added to the composition, they can be added in amounts known in the art for their intended application or in somewhat lower doses than when administered alone.
The pharmaceutically acceptable salts must be pharmacologically and pharmaceutically or veterinarily acceptable, and can be prepared as alkaline or alkaline earth metal salts, such as sodium, potassium or calcium salts. Organic salts and esters are also suitable for use with this invention. The active compounds are preferably administered to a subject as a pharmaceutical or veterinary composition, which includes systemic and topical formulations. Among these, preferred formulations are suitable for inhalation, or for breathable, oral, oral, rectal, vaginal, nasal, intrapulmonary, ophthalmic, optical, intracavity, intratracheal, intraorganic, topical (including buccal, sublingual, dermal and intraocular) administration. , parenteral (including subcutaneous, intradermal, intramuscular, intravenous and intraarticular) and transdermal administration, among others. The present invention also provides a kit or kit comprising the composition and a device for administration. The compositions can be conveniently presented in single or multiple unit dosage form as well as in bulk, and can be prepared by any of the methods that are well known in the art of pharmacy. The composition, is in the kit, already formulated with or in which the first and second active agents are separately provided together with other ingredients, and the instructions for their formulation and administration regime. The kit can also contain other agents, such as those described in this patent, and for example, when for parenteral administration, they can be provided with a carrier in a separate container, wherein the carrier can be sterile. The present composition may also be provided in lyophilized form, and in a separate container, which may be sterile, to be added to a liquid carrier prior to administration. See, e.g. Patent No. 4,956,355; UK Patent No. 2,240,472; Patent Application EPO Series No. 429,187; PCT Patent Publication No. 91/04030; Mortensen, S.A., et al., Int. J. Tiss. Reac. XII (3): 155-162 (1990); Greenberg, S. et al., J. Clin. Pharm. 30: 596-608 (1990); Folkers, K., et al., Proc. Nati Acad. Sci. USA 87: 8931-8934 (1990), the relevant compound and preparatory portions of which are incorporated by reference above. The present composition is provided in a variety of systemic and topical formulations. The systemic or topical formulations of the invention are selected from the group consisting of oral, intrabuccal, intrapulmonary, rectal, intrauterine, intradermal, topical, dermal, parenteral, intratumoral, intracranial, intrapulmonary, buccal, sublingual, nasal, subcutaneous, intravascular , intrathecal, inhalable, breathable, intra-articular, intracavity, implantable, transdermal, iontophoretic, intraocular, ophthalmic, vaginal, optic, intravenous, intramuscular, intraglandular, intraorganic, intralymphatic, enteric coating formulations and slow release. The actual preparation and mixing of these different formulations are known in the art and do not need to be detailed in the present description. The composition can be administered once or several times a day. Formulations suitable for respiratory, nasal, intrapulmonary and inhalation administration are preferred, as are topical, oral and parenteral formulations. All methods of preparation include the step of carrying the active compounds in association with a carrier which constitutes one or more of the additional ingredients. In general, the formulations are prepared by bringing the active compound uniformly and intimately in association with a liquid carrier, a finely divided solid carrier, or both, and subsequently, if necessary, shaping the product into the desired formulations. Compositions suitable for oral administration can be presented in discrete units, such as capsules, cachets, pills or tablets, each containing a predetermined amount of the active compound; as a powder or granules; as a solution or a suspension, in an aqueous and non-aqueous liquid; or as an oil in water or water in oil emulsion. Compositions suitable for parenteral administration comprise sterile aqueous and non-aqueous injectable solutions of the active compound, which preparations are preferably isotonic with the blood of the proposed container. These preparations may contain antioxidants, buffers, antibacterials and solutes which make the compositions isotonic with the blood of the proposed container. Aqueous and non-aqueous sterile suspensions may include suspending agents and thickening agents. The compositions may be presented in unit dose or multi-dose containers, for example small vials and sealed vials, and may be stored in a freeze-dried or frozen-dry condition that requires only the addition of the sterile liquid carrier, e.g., saline solution. or water to inject immediately before use. Nasal and instillable formulations comprise purified aqueous solutions of the active compound with preservatives and isotonic agents. Such formulations are preferably adjusted to a pH and isotonic state compatible with the nasal mucous membranes. Formulations for rectal or vaginal administration may be presented as a suppository with a suitable carrier such as cocoa butter or hydrogenated fats or hydrogenated fatty carboxylic acids. Ophthalmic formulations are prepared by a method similar to nasal spray except that the pH and isotonic factors are preferably adjusted to match those of the eye. Otic formulations are generally prepared in viscous carriers, such as oils and the like, as is known in the art, such that they can be easily administered into the ear without spilling. Compositions suitable for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil. Carriers that may be used include Vaseline, lanolin, polyethylene glycols, alcohols, transdermal enhancers, and combinations of two or more thereof. Compositions suitable for transdermal administration can be presented as discrete patches adapted to remain in intimate contact with the epidermis of the container for a prolonged period of time.
The first and second active agents described herein can be administered to the respiratory system either by inhalation, respiration, nasal administration or intrapulmonary instillation (in the lungs) of a subject by any suitable means, and are preferably administered by generating an aerosol or spray that It comprises nasal, powder, or liquid particles, intrapulmonary, respirable or inhalable. The respirable or inhalable particles comprising the active compound are inhaled by the subject, i.e., by inhalation or by nasal administration or by instillation into the respiratory tract or only into the lung. The formulation may comprise respirable or breathable solid or liquid particles of the active compound which, in accordance with the present invention, include respirable or inhalable particles of a size small enough to pass through the mouth and larynx with inhalation and continue to the bronchi and alveoli of the lungs. In general, the particles vary from about 0.05, about 0.1, about 0.5, about 1, about 2 to about 4, about 6, about 8, about 10 microns in diameter. More particularly, about 0.5 to less than about 5um in diameter, are respirable or inhalable. Particles of non-respirable size that are included in an aerosol or dew tend to settle in the throat and be swallowed. The amount of non-respirable particles in the aerosol is thus, preferably minimized. For nasal administration or intrapulmonary instillation, a particle size in the range of about 8, about 10, about 20, about 25 to about 35, about 50, about 100, about 150, about 250, about 500 um (diameter) is preferred. to ensure retention in the nasal cavity or for instillation and direct deposition in the lung. Liquid formulations can be introduced by syringe or jets into the respiratory tract (nose) and lung, particularly when administered to newborns and infants. Liquid pharmaceutical compositions of the active compound for producing an aerosol can be prepared by combining the active compound with a stable vehicle, such as sterile, pyrogen-free water. The solid particulate compositions containing respirable dry particles of the micronized active compound can be prepared by grinding the dry active compound with a mortar and pestle, and then passing the micronized composition through a 400 mesh screen to break up or separate the agglomerates. big. A solid particulate composition comprised of the active compound may optionally contain a dispersant which serves to facilitate the formation of an aerosol. A suitable dispersant is lactose, which can be mixed with the active compound in any suitable ratio, e.g., a weight ratio of 1 to 1. U.S. Patent Applications. Serial Nos. 10 / 462,901 and 10 / 462,927 describe a stable dry powder formulation of DHEA in a nebulizable form and a stable dry powder formulation in the form of DHEA-S dihydrate crystals, respectively (these patent applications are incorporated herein) in its entirety by reference). The aerosols of liquid particles comprising the active compound can be produced by any suitable means, such as with a nebulizer. See, e.g. U.S. Pat. No. 4,501,729 (the description of which is incorporated herein by reference). Nebulizers are commercially available devices which transform solutions or suspensions of the active ingredient into a therapeutic aerosol mist either by accelerating a compressed gas, typically air or oxygen, through a narrow venturi or by means of agitation ultrasonic Compositions suitable for use in a nebulizer consist of the active ingredient in liquid carrier, the active ingredient comprises up to 40% w / w of the composition, but preferably less than 20% w / w of the carrier which is typically water or a dilute aqueous alcohol solution, preferably made isotonic with body fluids by the addition of, for example sodium chloride. Optional additives include preservatives if the composition is not prepared sterile, for example methyl hydroxybenzoate, antioxidants, flavoring agents, volatile oils, buffering agents and surfactants. The aerosols of solid particles comprising the active compound can also be produced with any aerosol generator of the sold particulate medicament. Aerosol generators for administering drugs in solid particles to a subject produce particles that are respirable, as explained above, and generate an aerosol volume containing a predetermined metered dose of a medicament at a rate suitable for human administration. Examples of such aerosol generators include metered dose inhalers and insufflators. The composition can be supplied with any delivery device that generates aerosols in solid or liquid particles, such as aerosol or spray generators. These devices produce respirable particles, as explained above, and generate a volume of aerosol or spray containing a predetermined metered dose of a drug at a rate suitable for human or animal administration. An illustrative type of aerosol or solid particle spray generator is an insufflator, which is suitable for administering finely ground powders. In the insufflator, the powder, e.g. A measured dose of the composition effective to carry out the treatments described herein is contained in a capsule or cartridge. These capsules or cartridges are typically made of gelatin, metal foil or plastic and can be perforated or opened in situ and the powder supplied by entraining air through the device with inhalation or by means of a manually operated pump. The composition employed in the insufflator may consist of either only the first and second active agents or a powder mixture comprising the first and second agents, typically comprising 0.01 to 100% w / w of the. composition, the composition generally contains the first and second agents in an amount of about 0.01% w / w r about 1% w / w, about 5% w / w, at about 20% w / w, about 40% w / w , approximately 99.9% p / p. Other ingredients, and other amounts of the agent, however, are also suitable within the limits of this invention.
In one embodiment, the composition is delivered by a nebulizer. This medium is specifically useful for patients or subjects who are unable to inhale or breathe the composition under their own efforts. In serious cases, patients or subjects are kept alive through an artificial respirator. The nebulizer can use any pharmaceutically or veterinarily acceptable carrier, such as a weak saline solution. The nebulizer is the means by which the powdered pharmaceutical composition is delivered to the target of patients or subjects in the airways. The composition is also provided in various forms that are adapted for different administration methods and delivery or delivery routes. In one embodiment, the composition comprises a breathable formulation, such as an aerosol or spray. The composition of the invention is provided in bulk and in unit form, as well as in the form of an implant, a capsule, blister or cartridge, which can be opened or perforated as is known in the art. Also provided is a kit, comprising a delivery device, and in separate containers, the composition of the invention, and optionally another excipient and therapeutic agents, and instructions for the use of the components of the kit. In one embodiment, the composition is delivered using a metered dose inhalation (MDI) formulation in suspension. Such an MDI formulation can be supplied using a delivery device using a propellant such as hydrofluoroalkane (HFA). Preferably, the HFA propellants contain 100 parts per million (PPM) or less of water.
In one embodiment, the delivery device comprises a dry powder inhaler (DPI) that delivers single or multiple doses of the composition. The single dose inhaler may be provided as a disposable case which is pre-sterilized with sufficient formulation for one application. The inhaler can be provided as a pressurized inhaler and the formulation in a capsule or cartridge that can be opened or punctured. The kit may also optionally comprise in a separate container an agent such as other therapeutic compounds, excipients, surfactants, (proposed as therapeutic agents as well as formulation ingredients), antioxidants, flavoring and coloring agents, fillers, volatile oils, buffering agents, dispersants, surfactants, antioxidants, flavoring agents, non-digestible materials, propellants and preservatives, among other additives suitable for the different formulations. Having described the present invention now in a general form, it will be understood by reference to certain specific examples, which are included herein for purposes of illustration only and are not intended to be limiting of the invention or of any embodiment of the invention, unless be specified.
EXAMPLES Examples 1 and 2: Live I Effects of Folinic Acid and DHEA in Adenosine Levels A 344 male young adult rats (120 grams) were administered dehydroepiandrosterone (DHEA) (300 mg / kg) or methyltestosterone (40 mg / kg) in carboxymethyl cellulose by priming once a day for fourteen days. Folinic acid (50 mg / kg) was administered intraperitoneally once a day for fourteen days. On day fifteen, animals were sacrificed by microwave pulses (1.33 kilowatts, 2450 megahertz, 6.5 seconds (s)) to the skull, which instantly denatures the entire brain protein and also prevents the metabolism of adenosine. The hearts of the animals were separated and instantly frozen in liquid nitrogen with 10 s of death. The liver and lungs were separated en bloc and were frozen instantaneously with 30 s of death. Brain tissue was subsequently dissected. Tissue adenosine was extracted, derivatized with 1, N6-ethenoadenosine and analyzed by high performance liquid chromatography (HPLC) using spectrofluorometric detection according to the method of Clark and Dar (J. of Neuroscience ethods 25: 243 (1988)) . The results of these experiments are summarized in the following Table 1. The results are expressed as the mean + SEM, with p <0.05 compared to the control group., And? p < 0.05 compared to DHEA or groups treated with methyltestosterone. Table 1: In vivo effect of DHEA, d-1-methyltestosterone and Folinic Acid on Adenosine Levels in various intracellular Adenosine Rat Tissues (nmol) / mg protein Heart Liver Lung Brain Treatment Control 10.6 + 0.6 14.5 + 1.0 3.1 + 0.2 0.5 + 0.04 DHEA. 6.7 + 0.5 16.4 ± 1.4 2.3 + 0.3 0.19 0.01 Methyltestosterone 8.3 ± 1.0 16.5 + 0.9 N.D. 0.42 + 0.06 (40 mg / kg) (? = 6)? (? = 6)? (? = 6)? Methyltestosterone 6.0 ± 0.4 5.1 + 0.5 N.D. 0.32 ± 0.03 Folinic Acid 12.4 + 2.1 16.412.4 N.D. 0.7210.09 DHEA (300 mg / kg) + 11.1 + 0.6 18.8 + 1.5 N.D. 0.5510.09 Folinic acid (50 mg / kg) Methyltestosterone 9.110.4 N.D. N.D. 0.60 + 0.06 (120 mg / kg) + Folinic Acid (50 mg / kg) N.D. = Not determined The results of these experiments indicate that the rats given DHEA or methyltestosterone daily for two weeks showed a depletion of adenosine in multiple organs. Depletion was dramatic in the brain (60% depletion for DHEA, 34% for methyltestosterone in high doses) and in the heart (37% depletion for DHEA, 22% depletion for high-dose methyltestosterone). Coadministration of folinic acid completely abolished steroid-mediated adenosine depletion. The folinic acid administered alone induced an increase in adenosine levels for all the organs studied.
Example 3: Anhydrous DHEA-S Air Jet Milling and Determination of Breathable Doses DHEA-S was evaluated as an asthma therapy. The solid state stability of sodium dehydroepiandrostenone sulfate (NaDHEA-S) has been studied for both the volumetric material and the ground material (Nakagawa, H., Yoshiteru,., And Fujimoto, Y. (1981) Chem. Phar. Bull., 29 (5) 1466-1469, Nakagawa, H., Yoshiteru, T., and Sugimoto, I. (1982) Chem. Pharm. Bull. 30 (1) 242-248). DHEA-S is very stable and crystalline like the dihydrate form. The anhydrous form of DHEA-S has low crystallinity and is very hygroscopic. The anhydrous form of DHEA-S is stable while it is collected without water in storage. The maintenance of a partially crystalline moisture-free material requires specialized manufacturing and packaging technology. For a solid product, it is essential to minimize sensitivity to moisture during the development process. (1) DHEA-S labeling The anhydrous DHEA-S was micronized using a jet mill (Jet-O-Mizer Series # 00, 100-120 PSI nitrogen). Approximately 1 g of sample was passed through the jet mill twice. The particles from each grind run were suspended in hexane, in which DHEA-S was insoluble and a Spa85 surfactant was added to prevent agglomeration. The resulting solution was sonicated for 3 minutes and seemed to completely disperse. The dispersed solutions were evaluated in a Malvern Mastersizer X apparatus with a low volume sampling attachment (SVS). A sample of dispersed material was evaluated 5 times. The average particle size or D (v, 0.5) of unground material was 52.56 μp? and the% RSD (relative standard deviation) was 7.61 for the 5 values. The D (v, 0.5) for a single pass through the jet mill was 3.90 pm and the RSD% was 1.21, and the D (v, 0.5) of a double pass through the jet mill was 3.25 im and the% of RSD was 3.10. This shows that DHEA-S can be jet-milled to particles of adequate size for inhalation. (2) HPLC analysis Two small flasks (A, single pass, 150 mg) and (B, double pass, 600 mg) of the micronized drug were available to determine the degradation of the drug during the micronization of jet milling. Weighted aliquots of DHEA-S from small vials A and B were compared with a standard unbleached DHEA-S solution (10 mg / ml) in a solution of acetonitrile-water (1: 1). The chromatographic peak area for the HPLC assay of the standard solution of the unmilled drug (10 mg / ml) gave a value of 23,427. Weighed aliquots of micronized DHEA-S were prepared (5 mg / ml) from the small bottles A and B, in a solution of acetonitrile-water (1: 1). The chromatographic peak areas for small bottles A and B were 11, 979 and 11,677, respectively. Clearly, there was no detectable degradation of the drug during the micronization process from jet milling. (3) Dose Studies Issued DHEA-S powder was collected in Nephele tubes and assayed by HPLC. Experiments were performed in triplicate at each air flow rate for each of the three dry powder inhalers tested (Rotahaler, Diskhaler and IDL DPI devices). A Nephele tube was fixed at one end with a glass filter (Gelman Sciences, Type A / E, 25 pm), which in turn was connected to the air flow line to collect the emitted dose of the drug from the respective dry powder inhaler that is tested. A silicone adapter was secured, with an opening to receive the mouthpiece of the respective dry powder inhaler that is tested at the other end of the Nephele tube. A desired air flow of 30, 60 or 90 L / min was achieved through the Nephele tube. Subsequently, each of the nozzles of the dry powder inhalers were inserted into the silicone rubber adapter, and the air flow was continued for approximately four seconds after which the tube was removed and the end cap was screwed on. the end of each tube. The end cap of the tube containing no filter was removed and 10 ml of an HPLC-grade water-acetonitrile (1: 1) solution was added to the tube, the end cap was replaced, and the tube was shaken for 1-2 minutes. Then the end cap was removed from the tube and the solution transferred to a 10 ml plastic syringe equipped with a filter (Carneo 13N syringe Filter, Nylon, 0.22 μp?). An aliquot of the solution was filtered directly into a small HPLC vial for an assay of the last drug via HPLC. The emitted dose experiments were performed with micronized DHEA-S (approximately 12.5 or 25 mg) which was placed in either a gelatin capsule (Rotahaler) or a Ventodisk blister (Diskhaler and single dose DPI (IDL)). When the micronized DHEA-S (only a small bottle B was used), it was weighed to place it in the gelatin capsule or blister, a few aggregates of the micronized powder appeared. The results of the tests of the emitted dose carried out at an air flow rate of 30, 60 and 87.8 L / min, are shown in Table 2. Table 2 summarizes the results of the Rotahaler experiments at 3 speeds Different flow rates, from Diskhaler experiments at 3 different flow rates, and from multiple dose experiments at 3 different current speeds. Table 2. Comparison of Dose Issued from Three Devices Dust Inhalers Dry Different Inhaler Device Flow Rate Dose Issued (%) air (L / min) Rotahaler 87.8 73.2, 67.1, 68.7 Average 69.7 Rotahaler (2 ^ study) 87.8 16.0, 24.5, 53.9 Average 31.5 Diskhaler 87.8 65.7, 41.6, 46.5 Average 51.3 Diskhaler (2nd study) 87.8 57.9, 59.9, 59.5 Average 59.1 Multiple Dose IDL 87.8 71.3, 79.0, 67.4 Average 72.6 Multiple Dose IDL 87.8 85.7, 84.6, 84.0 (2 * study) Average 84.8 Rotahaler 60 58.1, 68.2, 45.7 Average 57.3 Diskhaler 60 63.4, 38.9, 58.0 Average 68.2 Multiple doses IDL 60 78.8, 83.7, 89.6 Average 84.0 Rotahaler 30 34.5, 21.2, 48.5 Average 34.7 Diskhaler 30 53.8, 53.4, 68.7 58.6 Multiple Dose IDL 30 78.9, 88.2, 89.2 Average 85.4 (4) Breathable Dose Studies Breathable dose studies (respirable fraction) were performed using a standard cascade-type sampler impactor (Anderson), which consists of an inlet cone (a pre-separator of the impactor was substituted here), 9 stages, 8 collection plates and a support filter within 8 aluminum stages held together by 3 elastic fasteners and O-ring seals with packing, where each stage of the impactor contains multiple precision drilled holes. When air is drawn through the sampler, multiple jets of air at each stage direct any of the airborne particles to the surface of the collection plate for that stage. The dimension of the jets is constant for each stage, but it is smaller in each subsequent stage. If a particle is impacted at any given stage it depends on its aerodynamic diameter. The range of particle sizes collected in each stage depends on the jet velocity of the stage, and the cutoff point of the previous stage. Any particle not collected in the first stage follows the air current around the edge of the plate to the next stage, where either it is impacted or passed on in the subsequent stage, and so on, until the velocity of the jet is enough for the impact. To prevent the particle from bursting during the cascade impactor test, the individual impactor plates were coated with a hexane-fat (high vacuum) solution (100: 1 ratio). As noted above, the particle size cut-off points in the impactor plates were changed at different air flow rates. For example, Stage 2 corresponds to a higher cutoff value of 6.2 μp particles? at 60 L / min, and greater than 5.8 μp particles? at 30 L / min, and stage 3 had a particle size cutoff value at 90 L / min, greater than 5.6 μp ?. Thus, similar cut-off particle values were preferably used at comparable air flow rates, ie ranging from 5.6 to 6.2 μp ?. The equipment recommended by the United States Pharmacopeia to test dry powder inhalers consists of a nozzle adapter (silicone in this case) attached to a glass neck or throat (50 ml modified round bottom flask) and a distal pharynx of glass (induction port) leading to the top of the pre-separator and to the Andersen sampler. The sample of the pre-separator includes washes of the nozzle adapter, glass throat, distal glass pharynx and pre-separator. 5 ml of acetonitrile: water solvent (1: 1 ratio) was placed in the pre-separator before performing the cascade impactor experiment, which were carried out in duplicate with 3 different dry powder inhaler devices and at 3 flow speeds. air, 30, 60 and 90 L / min. The drug collected in the cascade impactor plates was tested by HPLC, and a mass balance of the drug was carried out for each multiple dose cascade impactor experiment and Diskhaler which consists of determining the amount of the drug left in the blister. , the amount of drug remaining in the device (only Diskhaler), the non-respirable amount of the dose retained in the silicone rubber nozzle adapter, glass throat, distal glass pharynx and pre-separator, all combined in a sample, and the respirable dose, i.e., Stage 2 a through the plates of the filter impactor for air flow rates of 30 and 60 L / min, and Stages 1 through the plates of the filter impactor for experiments of 90 L / min. Table 3. Experiments on the Waterfall Impactor (90 L / min) Device Pre-Blister Dose Device Balance Inhaler separator (%) Breathable (%) Mass (%) (%) (%) Diskhaler 72.7 6.6 2.9 22.1 104.3 Diskhaler 60.2 10.1 2.4 13.3 86.0 Multiple doses 65.8 3.9 3.8 26.5 * to 100.0 Multiple doses 73.3 3.8 3.6 19.3 * to 100.0 Dosage 78.7 2.8 4.6 13.9 * to 100.0 multiple * 13 Dose 55.9 5.0 1.2 37.9 * to 100.0 multiple * 0 * a: the multiple dose device was not washed; as solvents they could attack SLA components. The percentage retention of the multiple dose device was obtained by difference. * b: Drug dried in oven for 80 minutes * c: Drug dried in oven for 20 hours.
Based on the results of emitted doses and cascade impactor experiments, the low values of the low respirable doses obtained in the cascade impactor experiments were due to the agglomerated drug particles, which could not be separated, even in the highest air flow rate tested. The agglomeration of the drug particles is a consequence of the static charge increase during the mechanical grinding process used for particle size reduction and that this situation is further compounded by subsequent moisture absorption of the particles. A micronization method that produces less static charge or a completely hydrated, less hygroscopic crystalline form of DHEA-S (i.e., dihydrate form) should provide a freer flowing powder with decreased agglomeration potential. Example 4: Spray Drying of DHEA-S Anhydrous and Determination of Breathable Dose (1) Drug Micronization 1.5 g of anhydrous DHEA-S was dissolved in 100 ml of ethanol.50% water to produce a 1.5% solution. The solution was spray-dried with a B-191 Mini Spray Dryer (Buchi, Flawil, Switzerland) with an inlet temperature of 55 ° C, outlet temperature of 40 ° C, at 100% vacuum cleaner, at 10% pump, nitrogen flow at 40 mbar and spray flow at 600 units. The spray dried product was suspended in hexane and a Span85 surfactant was added to reduce agglomeration. The dispersions were sonicated with cooling for 3-5 minutes to complete the dispersion and the dispersed solutions were tested on a Malvern Mastersizer X apparatus with a Low Volume Tester (SVS) fixation. It was found that the two batches of spray-dried material had average particle sizes of 5.07 ± 0.70 μp? and 6.66 ± 0.91 μp? The optical examination by an optical microscope of the dispersions of each batch confirmed that spray drying produced small, respirable particles. The average particle size was 2.4 μp? and 2.0] im for each lot, respectively. This demonstrates that DHEA-S can be spray dried to a particle size suitable for inhalation. (2) Breathable Dose Studies The experiments in the cascade-type impactor were conducted as described in Example 3. Four experiments were made in the cascade impactor, three with a multiple-dose device IDL and one with a Diskhaler, all at 90 (L / min) The results of the cascade impactor experiments are presented in the following Table 4. The anhydrous spray-dried material in these experiments produced a two-fold increase in the respirable dose compared to the micronized anhydrous DHEA-S It appears that spray drying obtained higher respirable doses compared to jet milling, however, the percentage of respirable dose was still low, which was probably the result of moisture absorption of the anhydrous form. Cascade Impactor with a Spray Dried Drug Product Example 5: Air Jet Milling of DHEA-S Dihydrate (DHEA-S · 2H20) and Determination of Breathable Dose (1) Recrystallization of DHEA-S Dihydrate. Anhydrous DHEA-S was dissolved in a boiling mixture of 90% ethanol / water. This solution was rapidly cooled in a dry ice / methanol bath to recrystallize the DHEA-S. The crystals were filtered, washed twice with cold ethanol, then dried in a vacuum desiccator at room temperature (RT) for 36 hours. During the drying process, the material was periodically mixed with a spatula to break up the large agglomerates. After drying, the material was passed through a 500 m sieve. (2) icronization and physicochemical test. The DHEA-S dihydrate was micronized with nitrogen gas in a jet mill at a venturi pressure of 40 PSI, a mill pressure of 80 PSI, power setting of 25 and a product feed rate of approximately .120 to 175 g /hour. The surface area was determined using five BET point analyzes developed with nitrogen as the adsorption gas (P / PQ = 0.05 to 0.30) using a TriStar Micromeritic surface area analyzer. Particle size distributions were measured by laser diffraction using the Micrometrics Saturn Digisizer analyzer where the particles are suspended in mineral oil with sodium dioctyl sodium sulfosuccinate as a dispersing agent. The water content of the drug substance was measured by Karl Fischer titration (Sc ott Titroline KF). Pure water was used as the standard and all relative standard deviations in triplicates are less than 1%. Powder was added directly to the titration medium. The physicochemical properties of DHEA-S dihydrate before and after micronization are summarized in Table 5. Table 5. Physicochemical properties of DHEA-S dihydrate before and after micronization.
The only significant change measured is in the particle size. There was no significant water loss or increase in impurities. The surface area of the micronized material is in accordance with an irregularly shaped particle having a median size of 3 to 4 microns. Micronization successfully reduces particle size to a suitable range for inhalation without changes measured in solid state chemistry. (3) Aerosolization of DHEA-S dihydrate. The single-dose Acu-Breathe device was used to evaluate the DHEA-S dihydrate. Approximately 10 gr of powder of the pure DHEA-S dihydrate was filled and sealed in foil blister. These blisters were operated on the 8-stage Andersen waterfall impactor at current speeds ranging from 30 to 75 L / min, with a double blow glass throat. Steps 1-5 of the Andersen impactor are rinsed together to obtain an estimate of the fine particle fraction. The combination of the drug collected from the multiple stages in one trial makes the method much more sensitive. The results for this series of experiments are shown in Figure 1. At all flow rates, the dihydrate produces a finer particle fraction than the virtually anhydrous material. Since the dihydrate powder is atomized using the single dose inhaler, it is quite reasonable to conclude that its aerosol properties are significantly better than the virtually anhydrous material. Higher crystallinity and stable moisture content are the factors that most likely contribute to the superior aerosol properties of the dihydrate. This unique feature of DHEA-S dihydrate has not been reported in any previous literature. Although the improvement in the efficiency of the aerosol of DHEA-S with the dihydrate form is important, the substance of the pure drug may not be the optimal formulation. The use of a carrier with a larger particle size typically improves the aerosol properties of the micronized drug substances. Example 6: Stability of DHEA-S Anhydrous and DHEA-S Dihydrate with and without Lactose The initial purity (Time = 0) was determined for anhydrous DHEA and for DHEA-S dihydrate by high pressure liquid chromatography (HPLC) . Both forms of DHEA-S were then mixed, either with lactose in a ratio of 50:50, or used as a pure powder and placed in small open glass jars, and kept at 50 ° C for up to 4 weeks. These conditions were used to subject the formulation to an effort in order to predict its long-term stability results. Small control vials containing only DHEA-S (anhydrous or dihydrate) were sealed and kept at 25 ° C for up to 4 weeks. Samples were taken and analyzed by HPLC also at 0, 1, 2 and 4 weeks to determine the amount of degradation, as determined by the formation of DHEA. After one week, the virtually anhydrous DHEA-S mixed with lactose (50% w / w, nominally) stored at 50 ° C in sealed small glass bottles acquires a brownish hue that is darker for the lactose mixture. This color change is accompanied by a significant change in the chromatogram as shown in Figure 1. The primary degradant is DHEA. Qualitatively from Figure 2, the amount of DHEA in the mixture is higher than the other two samples. To quantitatively estimate the percentage of DHEA in the samples, the area of the DHEA peak is divided by the total area of the DHEA-S and DHEA peaks (see Table 6). The highest decomposition rate for the mixture indicates a specific interaction between lactose and virtually anhydrous DHEA-S. In parallel with the increase in DHEA, the brown color of the powders in accelerated storage increased with time. Materials in accelerated storage become more cohesive over time as evidenced by agglutination during the weighting of the sample for chemical analysis. Based on these results, it is not possible to formulate virtually anhydrous DHEA-S with lactose. This is a considerable disadvantage since lactose is the most commonly used inhalation excipient for dry powder formulations. Continuing the virtually anhydrous form would mean limiting the formulations to a pure powder or compromising more extensive safety studies to use a novel excipient.
Table 6: Percentage of DHEA formed from anhydrous DHEA-S at 50 ° C In contrast to Figure 2, there is virtually no DHEA generated after storage for 1 week at 50 ° C (see Figure 3). In addition, the materials show no change in color. The moisture content of the DHEA-S dihydrate remains virtually unchanged after one week at 50 ° C. The water content after accelerated storage is 8.66% against an initial value of 8.8%. The% DHEA measured during the course of this stability program is shown in Table 7. Table 7: Percentage of DHEA formed from DHEA-S dihydrate at 50 ° C Comparing Figures 1 and 2 and Tables 6 and 7, it can be seen that the dihydrate form of DHEA-S is the most stable form to move towards further studies.
The superior compatibility of DHEA-S dihydrate with lactose with respect to that of virtually anhydrous material has not been reported in the patent or literature sought. The solubility of this substance is reported in the next section as a portion of the development work for a nebulizer solution. Example 7: Mixtures of DHEA-S / Lactose dihydrate, Determination of Breathable Dose and Stability (1) Mixture of DHEA-S / Lactose dihydrate. Equal weights of DHEA-S and lactose grade inhalation (Foremost Aero Fio 95) were mixed manually, then passed through a 500 μm sieve to prepare a pre-mix. The pre-mix was then placed in a BelArt Micro-Mili micro-mill with the remaining lactose to produce 10% w / w of DHEA-S mixture. The mixer was reinforced by wire to a variable voltage source to regulate the speed of the propeller. The mixer voltage was cycled through 30%, 40%, 45% and 30% of the total voltage for 1, 3, 1.5, and 1.5 minutes, respectively. The uniformity of the content of the mixture was determined by HPLC analysis. Table 8 shows the result of the content uniformity samples for this mixture. The target value is 10% weight / weight of DHEA-S. The content of the mixture is satisfactory due to the proximity to the objective value and the uniformity of the content. Table 8: Uniformity of the content for a mixture of DHEA-S dihydrate with lactose. (2) Aerosolization of the DHEA-S / Lactose dihydrate mixture. Approximately 25 mg of this powder was emptied and sealed in metallic foil blister and aerosolized using the single dose device at 60 L / min. Two blisters were used for each test and the results for the fine particle fraction (material in steps 1-5) are shown in Table 9. The aerosol results for this preliminarily powdered mixture are satisfactory for a water supply system. respiratory drug. Higher fine particle fractions are possible with optimization of the powder mix and the blister / device configuration. The full particle size distribution of Test 2 is shown in Table 10. This average diameter for the DHEA-S of this aerosol is -2.5 μp ?.
This diameter is smaller than the average diameter measured for the DHEA-S dihydrate micronized by laser diffraction. Irregularly shaped particles can behave aerodynamically as smaller particles since their larger dimension tends to align with the airflow field. Therefore, it is common to see a difference between the two methods. The diffraction measurements are a quality control test for the input material while the cascade impaction is a quality control test for the finished product. Table 9: Fine particle fraction of a lactose mixture in two different experiments Table 10: Particle size distribution of DHEA-S / Lactose atomized dihydrate mixture (3) Stability of DHEA-S / Lactose dihydrate mixture. This lactose formulation was also placed in the accelerated stability program at 50 ° C. The results", for the content of DHEA-S are in Table 11. The control is the mixture stored at room temperature. There is no trend in DHEA-S content with respect to time for any condition and all results are within the range of samples collected for the content uniformity test (see Table 11). In addition, there are no color changes or irregularities observed in the chromatograms. The mixture appears to be chemically stable. Table 11: Strain stability data submitted to the DHEA-S / lactose dihydrate mixture at 50 ° C.
Example 8: Formulation of the DHEA-S Nebulizer Solubility of DHEA-S. An excess of the DHEA-S dihydrate, prepared according to the "Recrystallization of the DHEA-S dihydrate (Example 5)" was added to the solvent medium and allowed to equilibrate for at least 14 hours with some periodic agitation. The suspensions were then filtered through a 0.2 micron syringe filter and diluted immediately for HPLC analysis. To prepare the refrigerated sample, the syringes and filters were stored in the refrigerator for at least one hour before being used. Inhalation of pure water can produce a cough stimulus. Therefore, it is important to add halide ions for a nebulizer formulation with NaCl being the salt most commonly used. Since DHEA-S is a sodium salt, NaCl could reduce solubility due to the common ion effect. The solubility of DHEA-S at room temperature (24-26 ° C) and refrigerated (7-8 ° C) as a function of the concentration of NaCl is shown in Figure 4. The solubility of DHEA-S is reduced with the NaCl concentration. Lowering the storage temperature reduces the solubility at all NaCl concentrations. The effect of temperature is weaker at high NaCl concentrations. In triplicate, the solubility at -25 ° C and 0% NaCl varies from 16.5-17.4 mg / mL with a relative standard deviation of 2.7%. At 0.9% of refrigerated NaCl, the variation in triplicate is 1.1-1.3 mg / mL with a relative standard deviation of 8.3%. The balance between DHEA-S in the solid and solution states is: NaDHEA-Ssóiido < • DHEA-S "+ Na + = [DHEA-S"] [Na +] / [NaDHEA-S] solid Since the concentration of DHEA-S in the solid is constant (ie, physically stable dihydrate), the equilibrium expression simplified: Ksp = [DHEA-S-] [Na +] Based on this assumption, a graph of the solubility of DHEA-S versus the reciprocal of the total sodium cation concentration is linear with a slope equal to Ksp. This is shown in Figures 5 and 6 for equilibrium at room temperature and refrigerated, respectively. Based on the correlation coefficients, the model is an adjustment for the data in both the ambient and refrigerated temperature where the equilibrium constants were 2236 and 665 mM2, respectively. To maximize solubility, the NaCl level needs to be as low as possible. The minimum content of halide ion for a nebulizer solution should be 20 mM or 0.12% NaCl. To estimate a concentration of DHEA-S for the solution, a temperature drop of 10 ° C is assumed in the nebulizer during use (i.e., 15 ° C). The interpolation between the equilibrium constants versus the reciprocal absolute temperature, the Ksp at 15 ° C would be -1316 mM2. Each mole of DHEA-S contributes one mole of the sodium cation to the solution, therefore: Ksp = [DHEA-S "] [Na +] = [DHEA-S-] [Na + + DHEA-S-] = [DHEA- S-] 2 + [Na +] [DHEA-S "] which is resolved for [DHEA-S-] using the quadratic formula. The solution for 20 mM Na + with a Ksp of 1316 mM2 is 27.5 mM DHEA-S "or 10.7 mg / mL, therefore, a DHEA-S solution of 10 mg / mL in 0.12% NaCl is selected as a good candidate formulation to progress in the additional test The estimate for this formula does not count for any concentration effect due to evaporation of water from the nebulizer The pH of a DHEA-S solution of 10 mg / mL with 0.12% NaCl ranges from 4.7 to 5.6 Although this could be an acceptable pH level for an inhalation formulation, the effect of using a 20 mM phosphate buffer is evaluated.The results of solubility at RT for buffered and non-buffered solutions are shown in Figure 7. The presence of a buffer in the formulation suppresses solubility, especially at low NaCl levels As shown in Figure 8, the solubility data for the buffered solution falls on the same equilibrium line as for the solution not damped The decrease in solubility with the buffer is due to the additional sodium cation content. Maximizing solubility is an important goal and buffering the formulation reduces solubility. In addition, Ishihora and Sugimoto ((1979) Drug Dev. Indust. Pharm. 5 (3) 263-275) did not show a significant improvement in the stability of NaDHEA-S at neutral pH.
Stability studies. A DHEA-S formulation of 10 mg / mL was prepared in 0.12% NaCl during a short-term solution stability program. The aliquots of this solution were emptied into small transparent glass jars and stored at RT (24-26 ° C) and at 40 ° C. Samples were checked daily for DHEA-S content, DHEA content, and appearance. For each time point, samples were extracted in duplicate and diluted from each small vial. The DHEA-S content with respect to the duration of this study is shown in Figures 9 and 10. In the accelerated condition, the solution shows a faster decomposition rate and became turbid after two days of storage. The solution stored at RT is more stable and a light precipitate is observed on the third day. The study was interrupted on day three. The decomposition of DHEA-S is accompanied by an increase in the DHEA content as shown in Figure 10. Because DHEA is insoluble in water, having only a small amount in the formulation creates a cloudy solution (accelerated storage). ) or a crystalline precipitate (environmental storage). This explains why earlier visual assessments of the solubility of DHEA-S severely underestimate the solubility of the compound: small amounts of DHEA could lead the experimenter to conclude that the solubility limit of DHEA-S has been exceeded. The solution should easily be stable on the day of reconstitution in a clinical trial. The following section describes the aerosol properties of this formulation. Nebulizer studies. The DHEA-S solutions are nebulized using a Pari ProNeb Ultra compressor and an LC Plus nebulizer. The schematic for the assembly of the experiment is shown in Figure 11. The nebulizer was filled with 5 ml of solution and the nebulization was continued until the output became visually insignificant (4¾ to 5 minutes). The nebulizer solutions were tested using a California Instruments AS-6 6-stage impactor with a USP throat. The impactor is run at 30 L / min, for 8 s to collect a sample after one minute of nebulization time. At all other times during the experiment, the aerosol is extracted through the pass-through collector at approximately 33 L / min. The collection apparatus, the nebulizer, and the impactor were rinsed with the mobile phase and assayed by HPLC. 5 ml of DHEA-S in 0.12% NaCl was used in the nebulizer. This volume was selected as the practical upper limit for use in a clinical study. The results for the first 5 misting experiments are shown below: Table 12. Results of nebulization studies with DHEA-S * Only liquid tested discharged from the nebulizer; It was not weighed before and after the atomization or rinsing of the complete unit Nebulizer # 1 was driven to dryness in about 5 minutes while Nebulizer # 2 took a little less than 4.5 minutes. In each case, the volume of the liquid remaining in the nebulizer is approximately 2 ml. This liquid is initially cloudy after it is removed from the nebulizer and afterwards it is rinsed in 3-5 minutes. Even after this time, the 10 mg / mL solutions appear to have a small amount of coarse precipitate in them. Fine air bubbles in the liquid appear to cause initial turbidity. DHEA-S appears to be surface active (i.e., promotes foam) and this stabilizes air bubbles within the liquid. The precipitate in 10 mg / mL solutions indicates that the solubility of the drug substance is exceeded in the nebulizer medium. Therefore, the additional nebulization experiments in Table 13 are run at lower concentrations. Table 13 presents additional data of "dose" linearity versus concentration of the solution. Table 13. Results of additional nebulizer experiments with DHEA-S.
Nebulizer # 3 took just under 4.5 minutes to reach dryness. The mass in the collector was plotted against the initial concentration of the solution in Figure 12. There is good linearity from 0 to 7.5 mg / mL after the amount collected seems to start stabilizing. Although the reduction of the solubility by cooling is included in the calculation of the 10 mg / mL solution, any effect of concentration on the drug and on the NaCl content was neglected. Therefore, it is possible for a precipitate to form via supersaturation of the nebulizer liquid. The data in Figure 12 and the observation of some particles in the 10 mg / mL solution after nebulization indicate that the highest solution concentration for a test of the clinical trial concept formulation is approximately 7.5 mg / mL. . A sample of the. Spray was extracted in a cascade impactor for particle size analysis. There is no detectable trend in the particle size distribution with the solution concentration or with the nebulizer number. The average particle size distribution for all nebulization experiments is shown in Figure 13. The aerosol particle size measurements are in accordance with published / reported results for this nebulizer (ie, mean diameter of ~ 2 μp? ). Although in vitro experiments show that a nebulizer formulation can deliver respirable DHEA-S aerosols, the formulation is unstable and takes 4-5 minutes of continuous nebulization. Therefore, a stable DPI formulation has significant advantages. The DHEA-S dihydrate was identified as the most stable solid state for a DPI formulation. An optimal nebulizer formulation is 7.5 mg / mL of DHEA-S in 0.12% NaCl for clinical trials of DHEA-S. The pH of the formulation is acceptable without a buffer system. The aqueous solubility of DHEA-S is maximized by minimizing the concentration of the sodium cation. The minimum sodium chloride levels without buffer achieves this objective. This is the highest concentration of the drug with 20 mM of IC ~ that will not precipitate during nebulization. This formulation is stable for at least one day at RT. Example 9: Preparation of the Experimental Model Cell cultures were obtained, HT-29 SF cells, representing a sub-line of HY-29 cells (ATCC, Rockville, Md.) And adapted to grow in a serum-free PC-1 medium. completely defined (Ventrex, Portland, ME). The mother cultures were maintained in this medium at 37 ° C (in a humidified atmosphere containing 5% C02). At confluence the cultures were plated again after dissociation using trypsin / EDTA (Gibco, Grand Island, NY) and re-fed every 24 hours. Under these conditions, the doubling time for HT-29 SF cells during logarithmic growth was 24 hours. Flow Cytometry 105 cells were plated in 60 mm plates in duplicate. For the analysis of the cell cycle distribution, the cultures were exposed to 0, 25, 50 or 200 μ? of DHEA. For analysis of the reversal of the cell cycle effects of DHEA, the cultures were exposed to either 0 or 25 μ? of DHEA, and the media were supplemented with MVA, CH, RN, VA plus CH, or MVA plus CH plus RN or were not supplemented. The cultures were trypsinized after 0, 24, 48 or 74 hours and fixed and stained using a modification of a procedure of Bauer et al., Cancer Res. 46, 3173-3178 (1986). Briefly, cells were harvested by centrifugation and resuspended in cold phosphate-buffered saline. Cells were fixed in 70% ethanol, washed, and resuspended in phosphate buffered saline. HE. then added 1 ml of hypotonic staining solution (50 pg / ml propidium iodide (Sigma Chemical Co.), 20 g / ml Rnase A (Boehringer Mannheim, Indianapolis, IN), 30 mg / ml polyethylene glycol, 0.1% Triton X-100 in 5 mM citrate buffer, and after 10 minutes at room temperature, 1 ml of isotonic staining solution (propidium iodide, polyethylene glycol, Triton X-100 in 0.4 M NaCl) and the cells were analyzed using a flow cytometer, equipped with doublet discrimination of pulse duration / pulse area (Becton Dickinson Immunocytometry Systems, San Jose, C7A). After calibration with fluorescent beads, a minimum of 2xl04 cells / sample were analyzed, the data were the total number of s displayed cells in each of the 1024 channels of increasing fluorescence intensity, and the resulting histogram was analyzed using the program of Cellfit analysis (Becton Dickinson). Effect of DHEA on Cell Growth Cells were plated 25,000 cells in 30 mm plates per quadruplicate, and after 2 days received 0, 12.5, 25, 50 or 200 μ? of DHEA. Cell number was determined at 0, 24, 48 and 72 hours later using a Coulter counter (model Z, Coulter Electronics, Inc. Hialeah, FL). DHEA (AKZO, Basel, Switzerland) was dissolved in dimethyl sulfoxide, the filter sterilized, and stored at -20 ° C until used. Figure 14 illustrates the inhibition of growth for HT-29 cells by DHEA. The points refer to the cell numbers, and the bars refer to SEM. Each data point was made in quadruplicate, and the experiment was repeated three times. When the SEM bars are not apparent, the SEM is smaller than the symbol. Exposure to DHEA resulted in a reduced cell number compared to controls after 72 hours in 12.5 μ ?, 48 hours in 25 or 50 μ ?, and 24 hours in 200 μ? of DHEA, indicating that DHEA produced a growth inhibition dependent on time and dose. Effect of DHEA on the Cell Cycle To examine the effects of DHEA on the cell cycle distribution, HT-29 SF cells were plated (105 cells / 60 mm dish), and 48 hours later treated with 0.25, 50 or 200 μ? of DHEA. Figure 15 illustrates the effects of DHEA on the cell cycle distribution in HT-29 SF cells. After 24, 48 and 72 hours, the cells were harvested, fixed in ethanol and stained with propidium iodide, and the DNA / cell content was determined by flow cytometric analysis. The percentage of cells in the Gi, S and G2M phases was calculated using the Cellfit cell cycle analysis program. The S phase is marked by a quadrangle for clarity. The representative histograms are shown from determinations in duplicate. The experiment was repeated three times. The cell cycle distribution in cultures treated with 25 or 50 μ of DHEA was not changed after the first 24 hours. However, when the exposure time of DHEA increased, the proportion of cells in the S phase progressively decreased, and the percentage of cells in the Gi, S and G2M phases was calculated using the Cellfit cell cycle analysis program. The S phase was marked by a quadrangle for clarity. Representative histograms were shown from the duplicate determinations. The experiment was repeated three times. The distribution of the cell cycle in cultures treated with 25 or 50 μ? of DHEA were not changed after the initial 24 hours. However, when the exposure time to DHEA increased, the proportion of cells in the S phase progressively decreased and the percentage of cells in the Gi phase increased after 72 hours. A transient increase in the G2M phase cells was apparent after 48 hours. Exposure to 200 μ? of DHEA produced a similar but faster increase in the percentage of cells in Gi and a decreased proportion of cells in the S phase after 24 hours, which continued through the treatment. This indicates that DHEA produced a Gi block in HT-29 SF cells in a time and dose dependent manner. Example 10. Inversion of the DHEA-mediated Effect on Growth and Investment of the Cell Cycle of Growth Inhibition Mediated by DHEA. Cells were plated as previously, and after 2 days received either 0 or 25 μ? of a medium containing DHEA supplemented with mevalonic acid ("MVA"; mM) squalene (SQ; 80 um), cholesterol (CH; 15 g / ml), MVA plus CH, ribonucleosides (RN; uridine, cytidine, adenosine, and guanosine) in final concentrations of 30 μm each), deoxyribonucleosides (DN, thymidine, deoxycytidine, deoxyadenosine and deoxyguanosine in final concentrations of 20 μ each). RN plus DN, or MVA plus CH plus RN, or a medium that was not supplemented. All compounds were obtained from Sigma Chemical Co. (St. Louis, MO). The cholesterol was solubilized in ethanol immediately before use. The RN and DN used at maximum concentrations showed no effect on growth in the absence of DHEA. Figure 16 illustrates the reversal of DHEA-induced growth inhibition in HT-29 SF cells. In A, the medium was supplemented with MVA 2 μ ?, SQ 80 μ ?, 15 g / ml CH, or MVA plus CH (MVA + CH) or was not supplemented (CON). In B, the medium was supplemented with a mixture of RN containing uridine, cytidine, adenosine, and guanosine in final concentrations of 30 μ each; a mixture of DN containing thymidine, deoxycytidine, deoxyadenosine and deoxyguanosine in final concentrations of 20 μ? each; RN plus DN (RN + DN); or MVA plus CH plus RN (MVA + CH + RN). Cell numbers were assessed before and after 48 hours of treatment, and growth of the culture was calculated as the increase in cell number during the 48 hour treatment period. The columns represent the percentage of cell growth of untreated controls; the bars represent SEM. The increase in cell number in untreated controls was 173, 370"6518. Each data point represents quadrupled plates of four independent experiments.Static analysis was performed using the Student's t-test? P < 0.01;? P < 0.001, compared to the treated controls Note that the supplements have little effect on cell growth in the absence of DHEA Under these conditions, the growth inhibition induced by DHEA was partially overcome by the addition of MVA as well as the addition of MVA The addition of SQ or CH alone had no such effect, suggesting that the cytostatic activity of DHEA is partly mediated by endogenous mevalonate depletion and the subsequent inhibition of the biosynthesis of a previous intermediate in the path of cholesterol that is essential for cell growth.In addition, partial growth reconstitution was found after the addition of RN as well as after the RN plus DN but not after the addition of DN, indicating that the depletion of both the mevalonate and nucleotide mixtures are involved in the inhibitory action of DHEA growth. However, none of the reconstitution conditions including the combined addition of MVA, CH, 'and RN completely overcome the inhibitory action of DHEA, suggesting either cytotoxic effects or possibly that additional biochemical pathways are involved. Inversion of the Effect of DHEA on the Cell Cycle HT-29 SF cells were treated with 25 FM of DHEA in combination with a number of compounds, including MVA, CH or RN, to test their ability to prevent the specific effects of the cell cycle of DHEA. DHEA. The cell cycle distribution was determined after 48 hours and 72 hours using flow cytometry. Figure 17 illustrates the reversal of DHEA induced disruption in HT-29 SF cells. Cells were plated (105 cells / 60 mm dish) and 48 hours later treated with either 0 or 25 FM of DHEA. The medium was supplemented with 2 MVA FM, 15 Fg / ml CH; a mixture of NR containing uridine, cytidine, adenosine, and guanosine in final concentrations of 30 FM; MVA plus CH (MVA + CH); or MVA plus CH plus RN (MVA + CH + RN) or was not supplemented. The cells were harvested after 48 or 72 hours, fixed in ethanol, and stained with propidium iodide., and the DNA content per cell was determined by flow cytometric analysis. The percentages of the cells in the Gi, S, and GzM phases were calculated using the Cellfit cell cycle profile analysis program. The S phase was marked by a quadrangle for clarity. Representative histograms were shown from duplicate determinations. The experiment was repeated twice. Note that the supplements had little effect on the progress of the cell cycle in the absence of DHEA. Increasing the exposure time, DHEA progressively reduced the proportion of cells in the S phase. Although the inclusion of MVA partially prevented this effect in the initial 48 hours but not after 72 hours, the addition of MVA plus CH was also able of partially preventing the depletion of the S phase in 72 hours, suggesting a requirement of both MVA and CH for cell progression during prolonged exposure. The addition of MVA, CH, and RN was apparently more effective in reconstitution, but still without restoring the percentage of S-phase cells to the value observed in untreated control cultures. The CH or RN only had very little effect in 48 hours and no effect in 72 hours. Morphologically, the cells responded to DHEA by acquiring a rounded conformation, which was prevented only by the addition of MVA to the culture medium. Some of the DNA histograms after 72 hours of DHEA exposure in FIG. 4 also shows the presence of a subpopulation of cells that apparently have a reduced DNA content. Since the HT-29 cell line is known to carry populations of cells that contain variable numbers of chromosomes (68-72; ATCC), this may represent a subset of cells that have segregated by transporting few chromosomes. Conclusions Examples 9-10 above provide evidence that in in vitro exposure of HT-29 SF human colon adenocarcinoma cells at DHEA concentrations known to deplete the endogenous mevalonate results in inhibition of growth and in the arrest of G and that the addition of MVA to the culture medium partly prevents these effects. DHEA produced effects with protein isoprenylation which were in many considerations similar to those observed for the specific inhibitors of 3-hydroxy-3-methyl-glutaryl-CoA reductase such as lovastatin and compactin. Unlike the direct inhibitors of mevalonate biosynthesis, however, DHEA measured its effects by progressing the cell cycle and cell growth in a pleiotropic fashion involving the biosynthesis of ribo- and deoxynucleotides and possibly other factors as well. Example 11: Dose Inhaler Measures Active Ingredient Objective by Action Ipratropium bromide 25.0 pg DHEA 400 mg Stabilizer 5.0 g Trichlorofluoromethane 23.70 mg Dichlorodifluoromethane 61.25 mg Example 12: Dose Inhaler Measures Active Ingredient Objective by Action Ipratropium bromide 25.0 pg DHEA-S 400 mg Stabilizer 7.5 μ? Trichlorofluoromethane 23.67 mg Dichlorodifluoromethane 61.25 mg Example 13: Dose Inhaler Measures Active Ingredient Objective by Action Tiotropium bromide 25.0 μ? DHEA 400.0 mg Stabilizer 15.0 μg Trichlorofluoromethane 23.56 mg Dichlorodifluoromethane 61.25 mg Example 14: Measured Dose Inhaler Active Ingredient Objective by Drive Tiotropium bromide 25 .0 g DHEA-S 400 .0 mg Stabilizer 15 .0 Trichlorofluoromethane 23. 56 mg Dichlorodifluoromethane 61. 25 mg In the following Examples 15-18, the first and second active agents are micronized and mixed in bulk with lactose in the proportions given above. The mixture was emptied into hard gelatin capsules or cartridges or into double packs of specifically constructed metal foil blister packs (Rotadisks blister packs, Glaxo®) to be administered by an inhaler such as the Rotahaler inhaler (Glaxo®) or in the Case of the blister pack with the Diskhaler inhaler (Glaxo®).
Example 15: Dry Powder Formulation of Measured Dose Active Ingredient / cartridge or blister Ipratropium bromide 72.5 μg DHEA 1.00 mg Lactose Ph. Eur. At 12.5 or 25.0 mg Example 16: Dry Powder Formulation of Measured Dose Active Ingredient / cartridge or blister Ipratropium bromide 72.5 g DHEA-S 1 mg Lactose Ph. Eur. At 12.5 or 25.0 mg Example 17: Dry Powder Formulation of Dose Measure Active Ingredient / cartridge or blister Tiotropium bromide 72.5] ig DHEA 1 mg Lactose Ph. Eur. At 12.5 or 25.0 mg Example 18: Formulation of Regulated Dose Dry Powder Active ingredient / cartridge or blister Tiotropium bromide 72.5 \ xq DHEA-S 1 mg Lactose Ph. Eur. to 12.5 or 25.0 mg Example 19: Effects of DHEA-S alone and in combination with muscarxin receptor blockade on EGF-induced ASM proliferation The following studies were aimed at determining whether DHEA sulfate regulates synthesis in human ASM cells that can lead to new therapeutic applications in the control of severe chronic asthma. Human ASM cells were obtained from the distal trachea of transplant donors in compliance with the Institutional Review Board of the University of Pennsylvania. Distal tracheae were dissected free of connective tissue and enzymatically digested to produce approximately 1 x 10 4 human ASM cells. Subsequently these cells were grown to confluence in 10% fetal bovine serum and inactivated in serum-free media containing 1% bovine serum albumin for 24 hours. Subsequently, cells were incubated with either diluent, PDGF (10 ng / ml), EGF (1 ng / ml) of thrombin (lU / ml) for 24 hours. After the initial incubation, the cells were then exposed to [3 H] -thymidine and the DNA synthesis was measured for 24 hours using thymidine incorporation. The cells were scraped and placed on filters which were then counted using a beta counter. All the experiments were repeated in a minimum of three human ASM cell lines. All conditions per experiment were run with six replicates. The analysis of the data is represented as mean ± standard error of the average and the statistical analysis was carried out using the Bonferroni-Dunn correction and ANOVA with a proven meaning with P < 0.05. EGF produced an 8-fold increase in ASM proliferation, which was further increased in the presence of 10 μ? of methylcholine (Mch). Although the atropine of the muscarinic antagonist alone has no effect on the EGF response, it reverses the effect of the Mch. DHEA-S was able to attenuate the EFG response, in the presence or absence of the combination of Mch and atropine. See figure 18 for the results. Without the intention of limiting to a mechanism of action, these results indicate that in vivo, in the presence of bronchoconstrictor tone of the tonic airways, both atropine and DHEA-S can work together to inhibit the proliferation of smooth muscle of the Airways and reconstruction. In addition to DHEA-sulfate, other suitable non-glucocorticoid steroids may be used as the first active agent, including, but not limited to, epiandrosterone and derivatives, analogs, and pharmaceutically acceptable salts thereof. For example, as the compound represented by Formulas I, III and IV described herein. Although the invention has been described with reference to the presently preferred embodiments, it should be understood that various modifications may be made without departing from the spirit of the invention. All publications, patents and patent applications, and internet sites are hereby incorporated by reference in their entirety to the extent that whether each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference. In its whole.

Claims (19)

  1. NOVELTY OF THE INVENTION Having described the invention as above, property is claimed as contained in the following: CLAIMS 1. A pharmaceutical composition comprising a pharmaceutically or veterinarily acceptable carrier, a first active agent and a second effective active agent for treating asthma, chronic obstructive pulmonary disease or a respiratory or pulmonary disease, (a) the first active agent is at least one non-glucocorticoid steroid selected from a non-glucocorticoid steroid that has the chemical formula and a non-glucocorticoid steroid of the chemical formula wherein Ri, R2, R3, I, Rs, R ?, s, B.9r io, R12, R13, R1 and R19 are independently H, OR, halogen, Ci_io alkyl or C1-10 alkoxy, R5 and R11 are independently OH, SH, H, halogen, pharmaceutically acceptable ester, pharmaceutically acceptable thioester, pharmaceutically acceptable ether, pharmaceutically acceptable thioether, pharmaceutically acceptable inorganic esters, monosaccharide, disaccharide or oligosaccharide pharmaceutically acceptable, spiro-oxirane, spirothirane, -OS02R20I -OPOR20R21 or Ci-10 alkyl, R5 and 6 taken together are = 0, Rio and R11 taken together are = 0; Ri5 is (1) H, halogen, C1-C10 alkyl, or Ci-Ci0 alkoxy when? 5 is -C (0) 0R22, (2) H, halogen, OH or C1-C10 alkyl when Ri6 is halogen , OH, or C 1 -C 10 alkyl, (3) H, halogen, C 1 -C 10 alkyl, C 1 -C 10 alkenyl, C 1 -C 10 alkynyl, formyl, C 1 -C 10 alkanoyl or epoxy when R 16 is OH, (4) OR, SH, H, halogen, pharmaceutically acceptable ester, pharmaceutically acceptable thioester, pharmaceutically acceptable ether, pharmaceutically acceptable thioether, pharmaceutically acceptable inorganic esters, pharmaceutically acceptable monosaccharide, disaccharide or oligosaccharide, spiro-oxirane, spirothirane, -OSO2R20 or - OPOR20R21 when Ri6 is H, or R15 and Rie taken together are = 0; Ri7 and Ría are independently (1) H, -OH, halogen, Ci-C10 alkyl or C1-10 alkoxy when R6 is H, OR, halogen, C1-C10 alkyl or -C (0) OR22 (2) H, | (C1-C10 alkyl) -amino, (Ci-Cio alkyl) n amino- (Ci-Cio alkyl), Ci-Cio alkoxy, Ca-Cio hydroxy-alkyl, Ci-Ci0-C1-C10alkyl, (Ci_C10) Ci-Ciokyloxy (halogen) malkyls, formyl, C1-C10 carbalkoxy or C1-C10 alkanoyloxy when R15 and Ri6 taken together are = 0, (3) Rn and Rie taken together are = 0; (4) R17 or R18 taken together with the carbon to which they are attached form a 3 to 6 member ring containing 0 or 1 oxygen atom; or (5) R15 and R17 taken together with the carbons to which they are attached form an epoxide ring; R20 and R21 are independently OH, pharmaceutically acceptable ester or pharmaceutically acceptable ether; R22 is H, (halogen) m- (C1-10 alkyl) or C1-10 alkyl; n is 0, 1 or 2; and m is 1, 2 or 3; or pharmaceutically or veterinarily acceptable salts thereof; and (b) the second active agent is an anticholinergic bronchodilator.
  2. 2. The pharmaceutical composition according to claim 1, characterized in that the first active agent is a non-gulcocorticoid steroid having the chemical formula where the broken line represents a single or double bond; R is hydrogen or a halogen; H in position 5 is present in the alpha or beta configuration or the compound of chemical formula I comprises a racemic mixture of arabas configurations; and ¾ is hydrogen or a multivalent organic or inorganic dicarboxylic acid covalently linked to the compound.
  3. 3. The pharmaceutical composition according to claim 1, characterized in that the first active agent is a non-glucocorticoid spheroid having the chemical formula (I), wherein the multivalent organic dicarboxylic acid is SO2O, phosphate or carbonate, wherein M comprises a counterion, wherein the counter ion is H, sodium, potassium, magnesium, aluminum, zinc, calcium, lithium ammonium, amine, arginine, lysine, histidine, triethylamine, ethanolamine, choline, triethanolamine, procaine, benzathine, tromethanin, pyrrolidine, piperazine , diethylamine, sulfatide * S020-C¾CHC¾OCOR3; I OCOR2 or phosphatide O I! "P-OCH2CHCH2OCOR.3, ¡! O OCOR2 wherein R and R, which may be the same different, are straight or branched C1-C1 alkyl glucuronide .
  4. The pharmaceutical composition according to claim 3, characterized in that the first active agent is dehydroepiandrosterone.
  5. 5. The pharmaceutical composition according to claim 3, characterized in that the first active agent is dehydroepiandrosterone sulfate.
  6. 6. The pharmaceutical composition according to claim 1, characterized in that the anticholinergic bronchodilator is an ipratropium or a tiotropium.
  7. The pharmaceutical composition according to claim 1, characterized in that it further comprises a ubiquinone or a pharmaceutically or veterinarily acceptable salt thereof, wherein the ubiquinone has the chemical formula wherein n is 1 to 12.
  8. The pharmaceutical composition according to claim 1, characterized in that the pharmaceutical composition comprises particles of respirable or breathable size.
  9. 9. The pharmaceutical composition according to claim 8, characterized in that the particles are approximately?. ??? at approximately 10 μp? of size .
  10. 10. The pharmaceutical composition according to claim 8, characterized in that the particles are from about 10 μm to about 100 μp? from 5 size.
  11. A kit comprising a delivery device and the pharmaceutical composition according to claim 1.
  12. 12. The kit according to claim 10, characterized in that the delivery device is an aerosol generator or a spray generator.
  13. The kit according to claim 12, characterized in that the aerosol generator comprises an inhaler.;
  14. 14. The kit according to claim 13, characterized in that the inhaler supplies individual pre-measured doses of the formulation.
  15. 15. The kit according to claim 13, characterized in that the inhaler comprises a nebulizer or insufflator.
  16. 16. A method for reducing the likelihood of or treating asthma in a subject, comprising administering to a subject in need of such treatment a prophylactically or therapeutically effective amount of the pharmaceutical composition of claim 1.
  17. 17. A method for reducing the likelihood of or treating chronic obstructive pulmonary disease in a subject, comprising administering to a subject in need of such treatment a prophylactically or therapeutically effective amount of the pharmaceutical composition of claim 1. 18. a method for treatment of a respiratory, pulmonary or malignant condition or condition, or to reduce the levels of, or sensitivity to, adenosine or adenosine receptors in a subject, which comprises administering to a subject in need of such treatment an amount prophylactically or The method according to claim 18, wherein the disorder or condition comprises asthma, chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), dyspnea, emphysema, respiration, asthma, pulmonary hypertension, pulmonary fibrosis, hypersensitive airways, level s increased adenosine or adenosine receptor, hypersensitivity to adenosine, infectious diseases, pulmonary bronchoconstriction, allergies or inflammation of the respiratory tract, depletion of ubiquinone or pulmonary surfactant, chronic bronchitis, bronchoconstriction, difficulty breathing, obstructed or impaired pulmonary routes , adenosine test for cardiac function, pulmonary vasoconstriction, impaired breathing, acute respiratory distress syndrome (ARDS), administration of adenosine or drugs that increase the level of adenosine, infant respiratory distress syndrome (childhood SDR), pain , allergic rhinitis, cancer or chronic bronchitis.
MXPA06001231A 2003-07-31 2004-07-30 Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with an anticholinergic bronchodilator for treatment of asthma or chronic obstructive pulmonary disease. MXPA06001231A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US49222603P 2003-07-31 2003-07-31
US10/698,079 US20050038004A1 (en) 2003-07-31 2003-10-29 Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with an anticholinergic bronchodilator for treatment of asthma or chronic obstructive pulmonary disease
PCT/US2004/025055 WO2005011615A2 (en) 2003-07-31 2004-07-30 Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with an anticholinergic bronchodilator for treatment of asthma or chronic obstructive pulmonary disease

Publications (1)

Publication Number Publication Date
MXPA06001231A true MXPA06001231A (en) 2006-05-15

Family

ID=34118920

Family Applications (1)

Application Number Title Priority Date Filing Date
MXPA06001231A MXPA06001231A (en) 2003-07-31 2004-07-30 Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with an anticholinergic bronchodilator for treatment of asthma or chronic obstructive pulmonary disease.

Country Status (7)

Country Link
US (1) US20050038004A1 (en)
EP (1) EP1651170A4 (en)
JP (1) JP2007509839A (en)
AU (1) AU2004261294B2 (en)
CA (1) CA2534354A1 (en)
MX (1) MXPA06001231A (en)
WO (1) WO2005011615A2 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2002303425A1 (en) * 2001-04-24 2002-11-05 Epigenesis Pharmaceuticals, Inc. Composition, formulations and kit for treatment of respiratory and lung disease with non-glucocorticoid steroids and/or ubiquinone and a bronchodilating agent
EP1511500A4 (en) * 2002-06-12 2009-04-29 Epigenesis Pharmaceuticals Llc Combination of anti-muscarinic agents and non-glucocorticoid steroids
US20050101545A1 (en) * 2003-07-31 2005-05-12 Robinson Cynthia B. Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with an anticholinergic bronchodilator for treatment of asthma or chronic obstructive pulmonary disease
US20050026884A1 (en) * 2003-07-31 2005-02-03 Robinson Cynthia B. Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a beta-agonist bronchodilator for treatment of asthma or chronic obstructive pulmonary disease
US9084799B2 (en) 2005-02-11 2015-07-21 Pulmagen Therapeutics (Synergy) Limited Inhaled combination therapy
DE602006004963D1 (en) 2005-02-11 2009-03-12 Argenta Discovery Ltd COMBINATION OF METHYLXANTHINE COMPOUNDS AND STEROIDS FOR THE TREATMENT OF CHRONIC RESPIRATORY DISEASES

Family Cites Families (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3505337A (en) * 1967-12-22 1970-04-07 Boehringer Sohn Ingelheim N - hydrocarbyl-substituted noratropinium,haloalkylates and o-acyl derivatives thereof
US4208423A (en) * 1978-11-24 1980-06-17 Syntex Inc. Anticholinergic bronchodilators
DE2903957A1 (en) * 1979-02-02 1980-08-07 Boehringer Sohn Ingelheim AGENT FOR TREATING NASAL HYPERSECRETION
US4393066A (en) * 1981-06-05 1983-07-12 Garrett David M Method for treatment of herpetic lesions
US4499064A (en) * 1982-06-03 1985-02-12 Clayton Foundation For Research Assessment of nutritional status of individuals
US4501729A (en) * 1982-12-13 1985-02-26 Research Corporation Aerosolized amiloride treatment of retained pulmonary secretions
US4575498A (en) * 1983-07-21 1986-03-11 Duke University Method for restoring depleted purine nucleotide pools
US4628052A (en) * 1985-05-28 1986-12-09 Peat Raymond F Pharmaceutical compositions containing dehydroepiandrosterone and other anesthetic steroids in the treatment of arthritis and other joint disabilities
NL194728C (en) * 1987-04-16 2003-01-07 Hollis Eden Pharmaceuticals Pharmaceutical preparation suitable for the prophylaxis or therapy of a retroviral infection or a complication or consequence thereof.
CH673459A5 (en) * 1987-05-15 1990-03-15 Eprova Ag
ES2043897T3 (en) * 1988-01-28 1994-01-01 Koeltringer Peter COMBINATION PREPARATION, ESPECIALLY FOR THE TREATMENT OF NERVOUS CELL DISEASES.
FR2631828B1 (en) * 1988-05-27 1994-05-20 Spiral Recherche Developpement USE OF A FOLINIC SUBSTANCE AS AN ANTI-AGGREGATE AGENT
US4931441A (en) * 1988-11-09 1990-06-05 Luitpold Pharmaceuticals, Inc. Stabilized aqueous leucovorin calcium compositions
US4920115A (en) * 1988-12-28 1990-04-24 Virginia Commonwealth University Method of lowering LDL cholesterol in blood
US5407684A (en) * 1988-12-30 1995-04-18 Virginia Commonwealth University Use of DHEA as a medicinal
US5077284A (en) * 1988-12-30 1991-12-31 Loria Roger M Use of dehydroepiandrosterone to improve immune response
IT1229517B (en) * 1989-01-31 1991-09-03 Bioresearch Spa USE OF 5-METHYLTETRAHYDROPHOLIC ACID, OF 5 FORMYLTHETRAHYDROPHOLIC ACID, AND OF THEIR PHARMACEUTICALLY ACCEPTABLE SALTS FOR THE PREPARATION OF PHARMACEUTICAL COMPOSITIONS IN THE FORM OF CONTROLLED RELEASE SUITABLE FOR BEING EMPLOYED IN THE TREATMENT OF DISORDERS IN THE TREATMENT OF DISORDERS.
IT1229203B (en) * 1989-03-22 1991-07-25 Bioresearch Spa USE OF 5 METHYLTHETRAHYDROPHOLIC ACID, 5 FORMYLTHETRAHYDROPHOLIC ACID AND THEIR PHARMACEUTICALLY ACCEPTABLE SALTS FOR THE PREPARATION OF PHARMACEUTICAL COMPOSITIONS IN THE FORM OF CONTROLLED RELEASE ACTIVE IN THE THERAPY OF MENTAL AND ORGANIC DISORDERS.
NL8901432A (en) * 1989-06-06 1991-01-02 Pharmachemie Bv STABLE AQUEOUS FOLINATE SOLUTION AT REFRIGERATOR TEMPERATURE, AND METHOD FOR PREPARING THAT.
US4985443A (en) * 1989-08-04 1991-01-15 Montes Leopoldo F Method and composition for treating vitiligo
US5173488A (en) * 1989-08-21 1992-12-22 American Cyanamid Company Stable injectable pharmaceutical formulation for folic acid and leucovorin salts and method
US5270305A (en) * 1989-09-08 1993-12-14 Glaxo Group Limited Medicaments
US5110810A (en) * 1991-02-08 1992-05-05 Virginia Commonwealth University Method of using dehydroepiandrosterone and dehydroepiandrosterone-sulfate as inhibitors of platelet aggregation
US5162198A (en) * 1991-02-08 1992-11-10 Virginia Commonwealth University Method of using dehydroepiandrosterone and dehydroepiandrosterone-sulfate as inhibitors of thrombuxane production and platelet aggregation
US5266312A (en) * 1992-01-07 1993-11-30 National Jewis Center For Immunology And Respiratory Medicine Method for treating a steroid resistant condition via administration of gamma interferon
WO1993016704A1 (en) * 1992-02-24 1993-09-02 East Carolina University Method of inhibiting carcinogenesis by treatment with dehydroepiandrosterone and analogs thereof
US6093706A (en) * 1992-03-04 2000-07-25 Bioresponse, L.L.C. Combined dehydroepiandrosterone and retinoid therapy for epithelial disorders
US5686438A (en) * 1993-03-09 1997-11-11 University Of Utah Research Foundation Methods for preventing progressive tissue necrosis, reperfusion injury, bacterial translocation and adult respiratory distress syndrome
AU6048694A (en) * 1992-12-09 1994-07-04 Paul D. Jager Stabilized medicinal aerosol solution formulations
AU5855794A (en) * 1993-01-19 1994-08-15 Endorecherche Inc. Therapeutic uses and delivery systems of dehydroepiandrosterone
US5635496A (en) * 1993-03-09 1997-06-03 University Of Utah Research Foundation Methods for preventing progressive tissue necrosis, reperfusion injury, bacterial translocation and adult respiratory distress syndrome
US5811418A (en) * 1993-03-09 1998-09-22 University Of Utah Research Foundation Methods for preventing progressive tissue necrosis, reperfusion injury, bacterial translocation and adult respiratory distress syndrome
HUT72968A (en) * 1993-03-09 1996-06-28 Univ Utah Res Found Process for producing pharmaceutical composition which is applicable for preventing progressive tissue necrosis, reperfusion injury, bacterial translocation and adult respiratory distress syndrome
US5407927A (en) * 1993-04-16 1995-04-18 The Regents Of The University Of California Treatment of mild depression and restoration of IGF-I levels in aging by dehydroepiandrosterone
EP0796112A4 (en) * 1994-11-30 1999-12-01 Amur Research Corp Phosphocholine drug derivatives
US20020032160A1 (en) * 1995-02-24 2002-03-14 Nyce Jonathan W. Compositions & formulations with an epiandrosterone or a ubiquinone & kits & their use for treatment of asthma symptoms & for reducing adenosine/adenosine receptor levels
US5660835A (en) * 1995-02-24 1997-08-26 East Carolina University Method of treating adenosine depletion
US5859000A (en) * 1995-06-07 1999-01-12 University Of Utah Research Foundation Method for reducing mast cell mediated allergic reactions
US5603918A (en) * 1995-06-09 1997-02-18 Boehringer Ingelheim Pharmaceuticals, Inc. Aerosol composition of a salt of ipratropium and a salt of albuterol
DE19528145A1 (en) * 1995-08-01 1997-02-06 Boehringer Ingelheim Kg New drugs and their use
US5767278A (en) * 1995-10-06 1998-06-16 Geron Corporation Telomerase inhibitors
EP0930876B2 (en) * 1996-07-22 2010-07-07 Renovo Limited Use of compounds that promote oestrogenic activity for acclerating the healing of skin wounds
US5861391A (en) * 1997-01-29 1999-01-19 Research Development Foundation Use of dehydroepiandrosterone to treat primary adrenal insufficiency and Addison's disease
US20030013772A1 (en) * 2000-02-23 2003-01-16 Murphy Michael A. Composition, synthesis and therapeutic applications of polyamines
DE19921693A1 (en) * 1999-05-12 2000-11-16 Boehringer Ingelheim Pharma Pharmaceutical composition for treating respiratory disorders, e.g. asthma, comprises combination of anticholinergic and beta-mimetic agents having synergistic bronchospasmolytic activity and reduced side-effects
DE60126597T2 (en) * 2000-04-28 2007-10-31 Inflazyme Pharmaceuticals, Ltd., Richmond 3-NITROGEN-6,7-DIOXYGENIZED STEROID COMPOUNDS AND USES THEREOF
DZ3477A1 (en) * 2000-10-12 2002-04-18
US7381713B2 (en) * 2000-12-04 2008-06-03 Sioan-Kettering Institute For Cancer Research Treatment of cancer by reduction of intracellular energy and pyrimidines
US20020189610A1 (en) * 2001-02-01 2002-12-19 Karl-Heinz Bozung Pharmaceutical compositions containing an ipratropium salt and a betamimetic
US6455028B1 (en) * 2001-04-23 2002-09-24 Pharmascience Ipratropium formulation for pulmonary inhalation
AU2002303425A1 (en) * 2001-04-24 2002-11-05 Epigenesis Pharmaceuticals, Inc. Composition, formulations and kit for treatment of respiratory and lung disease with non-glucocorticoid steroids and/or ubiquinone and a bronchodilating agent
US20030216329A1 (en) * 2001-04-24 2003-11-20 Robinson Cynthia B. Composition, formulations & kit for treatment of respiratory & lung disease with dehydroepiandrosterone(s) steroid & an anti-muscarinic agent(s)
AU2002303427A1 (en) * 2001-04-24 2002-11-05 East Carolina University Compositions and formulations with a non-glucocorticoid steroid and/or a ubiquinone and kit for treatment of respiratory and lung disease
US6608055B2 (en) * 2001-06-22 2003-08-19 Boehringer Ingelheim Pharma Kg Crystalline anticholinergic, processes for preparing it and its use for preparing a pharmaceutical composition
IL159238A0 (en) * 2001-06-22 2004-06-01 Boehringer Ingelheim Pharma Crystalline anticholinergic, method for its production, and use thereof in the production of a drug
US20030138434A1 (en) * 2001-08-13 2003-07-24 Campbell Robert L. Agents for enhancing the immune response
EP1511500A4 (en) * 2002-06-12 2009-04-29 Epigenesis Pharmaceuticals Llc Combination of anti-muscarinic agents and non-glucocorticoid steroids
US7405207B2 (en) * 2002-06-17 2008-07-29 Epigenesis Pharmaceuticals, Inc. Nebulizer formulations of dehydroepiandrosterone and methods of treating asthma or chronic obstructive pulmonary disease using compositions thereof
US20050101545A1 (en) * 2003-07-31 2005-05-12 Robinson Cynthia B. Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with an anticholinergic bronchodilator for treatment of asthma or chronic obstructive pulmonary disease

Also Published As

Publication number Publication date
US20050038004A1 (en) 2005-02-17
AU2004261294B2 (en) 2011-01-20
EP1651170A2 (en) 2006-05-03
AU2004261294A1 (en) 2005-02-10
AU2004261294A2 (en) 2005-02-10
CA2534354A1 (en) 2005-02-10
WO2005011615A3 (en) 2007-04-19
EP1651170A4 (en) 2009-04-01
JP2007509839A (en) 2007-04-19
WO2005011615A2 (en) 2005-02-10

Similar Documents

Publication Publication Date Title
US20050026850A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a tyrosine kinase inhibitor, delta opioid receptor antagonist, neurokinin receptor antagonist, or VCAM inhibitor for treatment of asthma or chronic obstructive pulmonary disease
AU2005232532B2 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a glucocorticosteroid for treatment of asthma, chronic obstructive pulmonary disease or allergic rhinitis
US20050085428A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a methylxanthine derivative for treatment of asthma or chronic obstructive pulmonary disease
MXPA06001162A (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a pde-4 inhibitor for treatment of asthma or chronic obstructive pulmonary disease.
US20050113317A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a cromone for treatment of asthma or chronic obstructive pulmonary disease
AU2004261294B2 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with an anticholinergic bronchodilator for treatment of asthma or chronic obstructive pulmonary disease
AU2004260700B2 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a beta-agonist bronchodilator for treatment of asthma or chronic obstructive pulmonary disease
US20050090455A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a lipoxygenase inhibitor for treatment of asthma or chronic obstructive pulmonary disease
US20080279789A1 (en) Combination Of Dehydroepiandrosterone Or Dehydroepiandrosterone-Sulfate With An Anticholinergic Bronchodilator For Treatment Of Asthma Or Chronic Obstructive Pulmonary Disease
US20050222049A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a glucocorticosteroid for treatment of asthma, chronic obstructive pulmonary disease or allergic rhinitis
US20090285898A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with an anticholinergic bronchodilator for treatment of asthma or chronic obstructive pulmonary disease
US20050113318A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a beta-agonist bronchodilator for treatment of asthma or chronic obstructive pulmonary disease
US20090297611A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a tyrosine kinase inhibitor, delta opioid receptor antagonist, neurokinin receptor antagonist, or vcam inhibitor for treatment of asthma or chronic obstructive pulmonary disease
US20090285900A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a beta-agonist bronchodilator for treatment of asthma or chronic obstructive pulmonary disease
US20090274676A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a pde-4 inhibitor for treatment of asthma or chronic obstructive pulmonary disease
US20050085430A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a PDE-4 inhibitor for treatment of asthma or chronic obstructive pulmonary disease
US20090317477A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a glucocorticosteroid for treatment of asthma, chronic obstructive pulmonary disease or allergic rhinitis
US20110209699A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a lipoxygenase inhibitor for treatment of asthma or chronic obstructive pulmonary disease
US20090285899A1 (en) Combination of dehydroepiandrosterone or dehydroepiandrosterone-sulfate with a methylxanthine derivative for treatment of asthma or chronic obstructive pulmonary disease

Legal Events

Date Code Title Description
FA Abandonment or withdrawal